[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (80)

Search Parameters:
Keywords = testicular injury

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 4971 KiB  
Article
Triptolide Causes Spermatogenic Disorders by Inducing Apoptosis in the Mitochondrial Pathway of Mouse Testicular Spermatocytes
by Jiantao Zhao, Maosheng Cao, Haisheng Yi, Guitian He, Tong Chen, Lingyun Liu, Kaimin Guo, Yin Cao, Chunjin Li, Xu Zhou, Boqi Zhang and Hongliang Wang
Toxics 2024, 12(12), 896; https://doi.org/10.3390/toxics12120896 - 10 Dec 2024
Viewed by 462
Abstract
Triptolide (TP) is a diterpenoid compound extracted from the traditional Chinese medicinal herb Tripterygium wilfordii. It has antitumor and anti-inflammatory effects and stimulates immunity. However, its serious side effects, especially reproductive toxicity, limit its clinical application. This study employed a testicular injury model [...] Read more.
Triptolide (TP) is a diterpenoid compound extracted from the traditional Chinese medicinal herb Tripterygium wilfordii. It has antitumor and anti-inflammatory effects and stimulates immunity. However, its serious side effects, especially reproductive toxicity, limit its clinical application. This study employed a testicular injury model established by intraperitoneally injecting TP (0.2 mg/kg) in C57BL/6J male mice (age = 7–8 weeks) for 14 days. The control and TP mice’s testicular tissues were subjected to transcriptome sequencing to assess potential testicular damage mechanisms. Based on the transcriptome sequencing results and relevant literature reports, further experiments were performed. In addition, to alleviate triptolide-induced testicular damage, we treated the mice with N-acetyl-L-cysteine (NAC). The acquired data revealed that compared with the control mice, the TP-treated mice’s testes indicated severe damage. Transcriptome sequencing identified differentially expressed genes that showed enrichment in cell differentiation, apoptotic process, cell cycle, glutathione (GSH) metabolism, and the p53 signaling pathway. Furthermore, TUNEL assays and Western blot analysis showed that in the TP mice’s testicular tissues, the spermatocytes had mitochondrial pathway apoptosis as well as abnormal mitochondrial morphology and structure. Triptolide induces oxidative stress in testicular tissue by enhancing pro-oxidative systems and inhibiting antioxidant systems. NAC reduced testicular damage and apoptosis by alleviating TP-induced oxidative stress. This study also employed a GC2 cell line for in-vitro analyses, and the results were consistent with the in vivo experiments. This study provides evidence for alleviating TP’s adverse effects on the male reproductive system for better clinical application. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Triptolide-induced testicular damage and spermatogenic disorders. (<b>A</b>) The chemical structure of triptolide. (<b>B</b>) A flowchart of medication administration. (<b>C</b>) The epididymal sperm count (n = 5). (<b>D</b>) The size of the testis (<b>a</b>) and organ index (<b>b</b>). (<b>E</b>) H&amp;E staining, the seminiferous tubule diameter, the epithelium height (<b>a</b>), and the tunica albuginea thickness of the testes (<b>b</b>). (<b>F</b>) The relative mRNA level of Sycp3 (n = 4). (<b>G</b>) The Western blot of Sycp3 (n = 3) (<b>a</b>,<b>b</b>). (<b>H</b>) The immunohistochemistry staining of testes. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control (means ± SEM).</p>
Full article ">Figure 2
<p>Transcriptomic analysis of testes. (<b>A</b>) A volcano plot of differential expression genes (DEGs). (<b>B</b>) The number of DEGs. (<b>C</b>) GO enhancement analyses of the DEGs. (<b>D</b>) KEGG enhancement analyses of the DEGs.</p>
Full article ">Figure 3
<p>Triptolide caused apoptosis of the mitochondrial pathway in testicular spermatogonia and GC2 cell lines. (<b>A</b>) TUNEL staining of testes. (<b>B</b>) Western blot analysis of Bax, Bcl-2, cleaved-caspase3 (n = 3) (<b>a</b>,<b>b</b>). (<b>C</b>) The cell viability of GC2 cells treated with different concentrations of triptolide. (<b>D</b>) The detection of apoptosis in GC2 cells by flow cytometry (<b>a</b>,<b>b</b>). (<b>E</b>) Western blot analysis of Bax, Bcl-2 (<b>a</b>), and cleaved-caspase3 (n = 3) (<b>b</b>). (<b>F</b>) Flow cytometric analysis was used to examine the mitochondrial membrane potential (MMP) levels in GC2 cells (<b>a</b>,<b>b</b>). (<b>G</b>) The electron microscopic observation of testicular (<b>a</b>) and GC2 cell mitochondria (<b>b</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control (means ± SEM).</p>
Full article ">Figure 4
<p>Triptolide led to oxidative stress in mouse testicular tissue and GC2 cell lines by enhancing the oxidative system and attenuating the antioxidant system (<b>A</b>) The MDA (<b>a</b>) and GSH (<b>b</b>) levels of testicular tissue. (<b>B</b>) The intracellular ROS levels of GC2 cell lines (<b>a</b>,<b>b</b>). (<b>C</b>) The MDA and GSH levels of GC2 cell lines (<b>a</b>,<b>b</b>). (<b>D</b>) The relative mRNA level of Gpx4, Txnd2, Hmox2, Aptx, Lcn2, Romo1 (<b>a</b>), and Cyp11a1 in testicular tissue (n = 4) (<b>b</b>). (<b>E</b>) The relative mRNA level of Gpx4, Hmox2, HO-1, Lcn2 (<b>a</b>), and Jun in GC2 cell lines (n = 3) (<b>b</b>). (<b>F</b>) The molecular docking of TP, Gpx4, Nrf2. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control (means ± SEM).</p>
Full article ">Figure 5
<p>The activation of oxidative stress-mediated apoptosis in the mitochondrial pathway by triptolide caused impaired spermatogenesis in mice. (<b>A</b>) A flowchart of medication administration. (<b>B</b>) The epididymal sperm count (n = 6). (<b>C</b>) The size of the testis (<b>a</b>) and the testis index (<b>b</b>). (<b>D</b>) H&amp;E staining (<b>a</b>), the seminiferous tubule diameter (<b>b</b>), the epithelium height (<b>c</b>), and the tunica albuginea thickness of testes (<b>d</b>). (<b>E</b>) The MDA (<b>a</b>) and GSH (<b>b</b>) levels of testicular tissue. (<b>F</b>) Western blot analysis of Bax, Bcl-2, cleaved-caspase3 of testicular tissues with different treatments (n = 3) (<b>a</b>–<b>e</b>). (<b>G</b>) TUNEL staining of the testes. (<b>H</b>) Relative mRNA levels of Sycp3 (n = 3). (<b>I</b>) Western blot analysis of Sycp3 of testicular tissues with different treatments (n = 3) (<b>a</b>,<b>b</b>). (<b>J</b>) Immunohistochemistry staining of the testes. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control (means ± SEM).</p>
Full article ">Figure 6
<p>The inhibition of oxidative stress alleviates triptolide-induced apoptosis of GC2 cell lines. (<b>A</b>) The cell viability of GC2 cells treated with different concentrations of NAC (1–5 mM). (<b>B</b>) The intracellular ROS levels of GC2 cell lines (<b>a</b>,<b>b</b>). (<b>C</b>) The MDA (<b>a</b>) and GSH (<b>b</b>) levels of GC2 cell lines. (<b>D</b>) Western blot analysis of Bax, Bcl-2, cleaved-caspase3 of GC2 cells with different treatments (n = 3) (<b>a</b>–<b>e</b>). (<b>E</b>) The detection of apoptosis in GC2 cells by flow cytometry (<b>a</b>,<b>b</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control (means ± SEM).</p>
Full article ">
24 pages, 6031 KiB  
Article
Protective Effects of L-Cysteine Against Cisplatin-Induced Oxidative Stress-Mediated Reproductive Damage
by Yi-Fen Chiang, Yi-Tzu Chen, Ko-Chieh Huang, Wei-Lun Hung, Cheng-Pei Chung, Tzong-Ming Shieh, Yun-Ju Huang, Mohamed Ali and Shih-Min Hsia
Antioxidants 2024, 13(12), 1443; https://doi.org/10.3390/antiox13121443 - 23 Nov 2024
Viewed by 469
Abstract
Cisplatin (CIS) is a widely used chemotherapeutic agent, but its side effects, such as oxidative stress, inflammation, and apoptosis, often lead to male reproductive damage. Oxidative stress, primarily caused by the excessive generation of reactive oxygen species (ROS), plays a critical role in [...] Read more.
Cisplatin (CIS) is a widely used chemotherapeutic agent, but its side effects, such as oxidative stress, inflammation, and apoptosis, often lead to male reproductive damage. Oxidative stress, primarily caused by the excessive generation of reactive oxygen species (ROS), plays a critical role in disrupting testicular homeostasis, resulting in spermatogenic impairment and tissue injury. L-cysteine (CYS), a semi-essential amino acid with potent antioxidant and anti-inflammatory properties, may offer protection against CIS-induced oxidative damage. This study aimed to assess the protective potential of CYS against CIS-induced male reproductive toxicity using in vivo and in vitro models. In vitro, treatment of TM3 (Leydig) and TM4 (Sertoli) cells with CIS led to increased ROS levels, reduced cell viability, and elevated apoptosis and inflammation, all of which were significantly ameliorated by subsequent CYS exposure. In vivo, CIS-treated male rats displayed heightened oxidative stress, impaired spermatogenesis, and histopathological damage in reproductive organs. However, CYS administration for 21 days significantly reduced oxidative stress, improved sperm viability, and protected testicular tissues from damage. These findings suggest that CYS has a protective effect against CIS-induced oxidative stress and male reproductive damage, making it a promising therapeutic agent for mitigating CIS-induced reproductive toxicity. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of L-cysteine (CYS) and cisplatin (CIS) on TM3 and TM4 cell viability. TM3 (5000 cells/well) (n = 7) and TM4 cells (3000 cells/well) (n = 7) were cultured in a 96-well plate for cell viability assays. Cells were treated with different concentrations of (<b>A</b>,<b>B</b>) CYS (0.1, 0.2, 0.5, 1, and 2 mM) or (<b>D</b>,<b>F</b>) CIS (5, 7.5, 10, 15, and 20 µM) for 24 or 16 h and stained with MTS assay. (<b>C</b>) TM3 and (<b>E</b>) TM4 cells (4 × 10<sup>4</sup> cell/well) were cultured in a 12-well plate for cell morphology assessment. Scale bar: 100 μm. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA following by Tukey’s multiple comparison test. *** <span class="html-italic">p</span> &lt; 0.001 compared with the control group.</p>
Full article ">Figure 2
<p>L-cysteine (CYS) attenuates cisplatin (CIS)-induced reduction in TM3 and TM4 cell viability. TM3 (5000 cell/well) (n = 7) and TM4 cells (3000 cell/well) (n = 7) were cultured in a 96-well plate for cell viability. TM3 (n = 7) and TM4 (n = 7) cells (4 × 10<sup>4</sup> cell/well) were cultured in a 12-well plate for cell morphology and composition. They were co-treated with different concentrations of CYS (0.1, 0.2, 0.5, 1 mM) and, respectively, induced by CIS (7.5 or 10 µM) for 24 and 16 h. (<b>A</b>) Morphology, (<b>B</b>,<b>C</b>) viability, and (<b>D</b>,<b>E</b>) crystal violet staining in TM3 and TM4 cells. Scale bar: 100 μm. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA following by Tukey’s multiple comparison test. *** <span class="html-italic">p</span> &lt; 0.001 compared with the control group. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 compared with CIS group.</p>
Full article ">Figure 3
<p>Effect of L-cysteine (CYS) on reactive oxygen species production in cisplatin (CIS)-induced TM3 and TM4 cells. The cells (1.2 × 10<sup>5</sup> cells/well) were cultured in a 6-well plate. They were co-treated with different concentrations of CYS (0.1, 0.2, 0.5, and 1 mM) and, respectively, induced by CIS (7.5 or 10 µM) for 24 and 16 h (n = 7). (<b>A</b>,<b>B</b>) ROS measurements were conducted by incubating the cells with 20 μM of DCFDA for 30 min at 37 °C in the dark. (<b>C</b>,<b>D</b>) HO-1 expression was analyzed by Western blot. (<b>E</b>) DPPH radical scavenging activity (%) of L-cysteine, gallic acid, and vitamin C. The IC50 DPPH values were obtained through extrapolation from regression analysis. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA followed by Tukey’s multiple comparison test. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 compared with the control group. # <span class="html-italic">p</span> &lt; 0.05 compared with the CIS group.</p>
Full article ">Figure 4
<p>Effect of L-cysteine (CYS) on apoptosis-related protein expression in cisplatin (CIS)-induced TM3 and TM4 cells. (<b>A</b>) TM3 and (<b>C</b>) TM4 cells (1 × 10<sup>5</sup> cells) were cultured in a 6 cm dish. Cells were co-treated with different concentrations of CYS (0.1, 0.2, 0.5, and 1 mM) and induced with CIS (7.5 or 10 µM) for 24 and 16 h, respectively (n = 4). The protein expression of PARP, Caspase 3, Bcl-2, and Bax was determined by Western blot, and the band values were normalized to GAPDH. (<b>B</b>,<b>D</b>) Quantification of protein expression. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA followed by Tukey’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared with the control group. # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 compared with the CIS group.</p>
Full article ">Figure 5
<p>Effect of L-cysteine (CYS) on testosterone secretion-related modulators in cisplatin (CIS)-induced TM3 cells. TM3 cells (1 × 10<sup>5</sup> cell/well) were cultured in a 6 cm dish. Cells were co-treated with different concentrations of CYS (0.1, 0.2, 0.5, 1 mM) and induced with CIS (7.5 µM) for 24 h (n = 4). (<b>A</b>) The inflammatory protein expressions were determined by Western blot. (<b>B</b>) iNOS and (<b>C</b>) COX2 protein expression levels were normalized to GAPDH. (<b>D</b>) Mitochondrial oxidative stress was evaluated by MitoSOX staining, with fluorescence intensity. (<b>E</b>) Testosterone secretion in the culture medium was quantified using an ELISA kit. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA followed by Tukey’s multiple comparison test. * <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 compared with the control group. # <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 compared with the CIS group.</p>
Full article ">Figure 6
<p>Effect of L-cysteine (CYS) on blood–testis barrier (BTB)-related protein expression in cisplatin (CIS)-induced TM4 cells. The cells (1 × 10<sup>5</sup> cell/well) were cultured in a 6 cm dish. Cells were co-treated with different concentrations of CYS (0.1, 0.2, 0.5, and 1 mM) and induced with CIS (10 µM) for 24 h (n = 4). (<b>A</b>) The BTB protein expressions were determined by Western blot. (<b>B</b>) ZO-1 and (<b>C</b>) occludin protein expression levels were normalized to GAPDH. Data are represented as mean ± SD. Different groups were analyzed using one-way ANOVA followed by Tukey’s multiple comparison test. *** <span class="html-italic">p</span> &lt; 0.001 compared with the control group. # <span class="html-italic">p</span> &lt; 0.05 compared with the CIS group.</p>
Full article ">Figure 7
<p>Effect of L-cysteine (CYS) on cisplatin (CIS)-induced weight and organ changes in in vivo model. (<b>A</b>) Body weight and (<b>B</b>) body weight changes, and the relative weight changes of the (<b>C</b>) testis, (<b>D</b>) epididymis, (<b>E</b>) heart, (<b>F</b>) liver, and (<b>G</b>) kidney in a CIS-induced animal model after CYS pretreatment (n = 8). Data are represented as mean ± SEM. Different groups were analyzed using Student’s <span class="html-italic">t</span>-test. *** <span class="html-italic">p</span> &lt; 0.001 compared with the CON group. # <span class="html-italic">p</span> &lt; 0.05 and ### <span class="html-italic">p</span> &lt; 0.001 compared with the CIS group. CON: control; CIS: cisplatin (10 mg/kg bw); LC: low CYS (100 mg/kg bw); HC: high CYS (300 mg/kg bw).</p>
Full article ">Figure 8
<p>Protective role of L-cysteine (CYS) treatment for the biochemical indicators in cisplatin-induced in vivo model. (<b>A</b>) Testosterone and P450scc protein expression were evaluated by Western blot; (<b>B</b>) SGOT, (<b>C</b>) SGPT, (<b>D</b>) BUN, (<b>E</b>) creatinine, (<b>F</b>) LDH, and (<b>G</b>) CPK levels in a cisplatin-induced animal model after CYS pretreatment (n = 8). Data are represented as mean ± SEM. Different groups were analyzed using Student’s <span class="html-italic">t</span>-test. * <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 compared with the CON group. # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 compared with the CIS group. CON: control; CIS: cisplatin (10 mg/kg bw); LC: low CYS (100 mg/kg bw); and HC: high CYS (300 mg/kg bw).</p>
Full article ">Figure 9
<p>Effect of L-cysteine (CYS) on testicular damage in cisplatin (CIS)-induced male rats. (<b>A</b>) The appearance of the testis and epididymis. (<b>B</b>) Hematoxylin-and-eosin-stained sections of the testis, observed under magnification at ×100, ×200, and ×400. Observations were made on multiple sections (2–3) from each testicular fragment to ensure consistent and representative assessment. Labels in the figure include Basement Membrane (BM, beige line), seminiferous tubules (STs, red line), Sertoli cells (SCs), sperm (S), spermatocytes (SPs, yellow circle), Interstitial Tissue (I), and Leydig cells (LCs). Treatment groups: CON (control), CIS (cisplatin, 10 mg/kg bw), LC (low CYS, 100 mg/kg bw), and HC (high CYS, 300 mg/kg bw).</p>
Full article ">Figure 10
<p>Effect of L-cysteine (CYS) on sperm morphology and viability in cisplatin (CIS)-induced animal model. (<b>A</b>) Sperm morphology stained with eosin Y and crystal violet, observed at 400× magnification. Black arrows indicate abnormal sperm, including (a) bent neck and (b) bent tail. (<b>B</b>) Sperm viability was assessed by counting the number of live and dead sperm. Data are represented as mean ± SEM. Different groups were analyzed using Student’s <span class="html-italic">t</span>-test. ** <span class="html-italic">p</span> &lt; 0.01 compared with the CON group. # <span class="html-italic">p</span> &lt; 0.05 compared with the CIS group. CON: control; CIS: cisplatin (10 mg/kg bw); LC: low CYS (100 mg/kg bw); and HC: high CYS (300 mg/kg bw).</p>
Full article ">
14 pages, 2475 KiB  
Article
PTD-FNK Alleviated LPS-Induced Oxidative Stress of Boar Testicular Sertoli Cells via Keap1-Nrf2 Pathway
by Weixia Ji, Qiuyan Huang, Qiqi Ma, Xingxing Song, Xin Zhang, Xun Li, Xiaoye Wang, Sutian Wang, Yanling Wang, Zhengzhong Xiao and Chuanhuo Hu
Vet. Sci. 2024, 11(11), 543; https://doi.org/10.3390/vetsci11110543 - 6 Nov 2024
Viewed by 1159
Abstract
PTD-FNK, a synthetic anti-apoptotic protein, has been shown to potently alleviate cellular injuries. However, the effects of PTD-FNK on oxidative defense in boar testicular Sertoli cells (SCs) against oxidative injury has not been explored. In this study, we show that exposure of SCs [...] Read more.
PTD-FNK, a synthetic anti-apoptotic protein, has been shown to potently alleviate cellular injuries. However, the effects of PTD-FNK on oxidative defense in boar testicular Sertoli cells (SCs) against oxidative injury has not been explored. In this study, we show that exposure of SCs to 100 mg/L lipopolysaccharide (LPS) for 12 h leads to decreased survival rate, superoxide dismutase (SOD) activity, and increased malondialdehyde (MDA). Treatment with 0.01 nmol/L PTD-FNK for 4 h significantly enhanced the activity of SOD, catalase (CAT), glutathione peroxidase (GSH-Px), and total antioxidant capacity (T-AOC) in SCs. Concurrently, PTD-FNK treatment effectively reduced the production of reactive oxygen species (ROS) and the levels of 8-hydroxy-2′-deoxyguanosine (8-OHdG) in SCs. Moreover, using His pull-down and LC-MS techniques, we identified PTD-FNK-interacting proteins and confirmed that this protective effect may be mediated by the regulation of the Keap1-Nrf2 signaling pathway by PTD-FNK. Therefore, PTD-FNK alleviates LPS-induced oxidative stress via the Keap1/Nrf2 pathway, providing novel insights for the development of therapeutic agents targeting testicular oxidative damage. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>,<b>B</b>) The morphology of SCs was identified by Oil Red O staining; (<b>C</b>) arrows indicate the bipolar corpuscles in the nucleus. The SC marker gene was detected by PCR. 1.<span class="html-italic">GATA4</span>; 2.<span class="html-italic">SOX9</span>; M.DL2000 Marker.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Effects of PTD-FNK on ROS production of LPS-induced boar SCs; (<b>C</b>) CCK8 assays of the PTD-FNK effect on cell viability; (<b>D</b>–<b>G</b>) effects of PTD-FNK on GSH-Px, CAT, 8-OHdG, and T-AOC contents of LPS-induced boar SCs; (<b>H</b>–<b>J</b>) effects of PTD-FNK on mRNA expression of GSH-Px, CAT, and SOD in LPS-induced boar SCs. <span class="html-italic">n</span> ≥ 3 (* <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).</p>
Full article ">Figure 3
<p>(<b>A</b>) His recombinant protein was detected by CBB; (<b>B</b>) His recombinant protein was detected by WB; (<b>C</b>) the expression of SCs His antibody was detected by WB; (<b>D</b>) “silver stain” detection result after pull-down; the red arrow represents the bait protein; the black arrows indicate the PTD-FNK interacting proteins; (<b>E</b>) Venn diagram of PTD-FNK differential protein sets in boar SCs; (<b>F</b>,<b>G</b>) PTD-FNK effects on <span class="html-italic">HSPA5</span> and <span class="html-italic">VIM</span> mRNA expression in boar SCs. <span class="html-italic">n</span> ≥ 3 ****<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>(<b>A</b>–<b>E</b>) The relative protein level of Nrf2, Keap1, HO-1, and <span class="html-italic">NQO1</span>; (<b>F</b>–<b>G</b>) Nrf2 and Keap1 protein levels were measured using ELISA; (<b>H</b>–<b>K</b>) <span class="html-italic">Nrf2</span>, <span class="html-italic">Keap1</span>, <span class="html-italic">HO-1</span>, and <span class="html-italic">NQO1</span> mRNA levels; control, no drug added; LPS, induced by 100 mg/L LPS for 12 h; LPS + PTD-FNK, protected by 0.01 mmol/L PTD-FNK for 4 h and then induced by 100 mg/L LPS for 12 h; LPS+ML385, induced by 5μM ML385 for 2 h, followed induced by 100 mg/L LPS for 12 h; LPS + PTD-FNK + ML385, induced by 5μM ML385 for 2 h, protected by 0.01 mmol /L PTD-FNK for 4 h, and then induced by 100 mg/L LPS for 12 h. <span class="html-italic">n</span> ≥ 3 (* <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).</p>
Full article ">
19 pages, 5333 KiB  
Review
Protective Role of Sphingosine-1-Phosphate During Radiation-Induced Testicular Injury
by Defan Wang, Renfeng Xu and Zhengchao Wang
Antioxidants 2024, 13(11), 1322; https://doi.org/10.3390/antiox13111322 - 30 Oct 2024
Viewed by 710
Abstract
The impact of ionizing radiation on the male reproductive system is gaining increasing attention, particularly when it comes to testicular damage, which may result in decreased sperm quality and hormonal imbalances. Finding effective protective measures to mitigate testicular damage caused by radiation has [...] Read more.
The impact of ionizing radiation on the male reproductive system is gaining increasing attention, particularly when it comes to testicular damage, which may result in decreased sperm quality and hormonal imbalances. Finding effective protective measures to mitigate testicular damage caused by radiation has become a focal point in the biomedical field. S1P, an essential biological signaling molecule, has garnered significant interest due to its multiple roles in regulating cellular functions and its protective effects against radiation-induced testicular injury. S1P not only effectively reduces the generation of ROS induced by radiation but also alleviates oxidative stress by enhancing the activity of antioxidant enzymes. Furthermore, S1P inhibits radiation-induced cell apoptosis by regulating the expression of anti-apoptotic and pro-apoptotic proteins. Additionally, S1P alleviates radiation-induced inflammation by inhibiting the production of inflammatory factors, thereby further protecting testicular tissue. In summary, S1P effectively reduces radiation-induced testicular damage through multiple mechanisms, offering a promising therapeutic approach to safeguard male reproductive health. Future research should explore the specific mechanisms of action and clinical application potential of S1P, aiming to contribute significantly to the prevention and treatment of radiation damage. Full article
(This article belongs to the Special Issue The Role of Oxidative Stress in Male Infertility)
Show Figures

Figure 1

Figure 1
<p>The testicular injury induced by radiation.</p>
Full article ">Figure 2
<p>The synthesis and metabolism of sphingosine 1-phosphate.</p>
Full article ">Figure 3
<p>The functions of sphingosine 1-phosphate signaling.</p>
Full article ">Figure 4
<p>Protective role of sphingosine 1-phosphate against radiation-induced testicular injury.</p>
Full article ">Figure 5
<p>The role of sphingosine 1-phosphate in reducing oxidative stress.</p>
Full article ">Figure 6
<p>The role of sphingosine 1-phosphate in inflammation regulation.</p>
Full article ">
16 pages, 5061 KiB  
Article
Polyethylene Terephthalate Microplastic Exposure Induced Reproductive Toxicity Through Oxidative Stress and p38 Signaling Pathway Activation in Male Mice
by Tianyang Li, Bohao Bian, Rihao Ji, Xiuwen Zhu, Xiaohui Wo, Qiankun Song, Zhigang Li, Feifei Wang and Yuqiao Jia
Toxics 2024, 12(11), 779; https://doi.org/10.3390/toxics12110779 - 25 Oct 2024
Viewed by 1148
Abstract
Polyethylene terephthalate (PET) is a type of polymer plastic that is often used to make plastic bags, bottles, and clothes. However, the waste of such plastic products is decomposed into microplastics (MPs), which are plastic fragments smaller than 5 mm, by various external [...] Read more.
Polyethylene terephthalate (PET) is a type of polymer plastic that is often used to make plastic bags, bottles, and clothes. However, the waste of such plastic products is decomposed into microplastics (MPs), which are plastic fragments smaller than 5 mm, by various external forces such as wind, UV radiation, mechanical wear, and biodegradation. PET MPs have been widely detected in the environment and human tissue samples; however, the toxicity and mechanism of PET MPs in mammals are still unclear. In this study, we investigated the male reproductive toxicity of PET MPs and their underlying mechanism. A total of 80 male mice were orally exposed to 0.01, 0.1, and 1 mg/d of PET MPs (with a diameter of 1 μm) for 42 days. The results showed that 1 μm PET MPs induced different degrees of pathological damage to testicular tissues, decreased sperm quality, and increased the apoptosis of spermatogenic cells via oxidative stress and p38 signaling pathway activation. To further illustrate and verify the mechanistic pathway, oxidative stress was antagonized using N-acetylcysteine (NAC), and the activation of the p38 signaling pathway was blocked using SB203580. The results revealed that the male reproductive injury effects after exposure to PET MPs were significantly ameliorated. Specifically, the testicular tissue lesions were relieved, the sperm quality improved, and the apoptosis of spermatogenic cells decreased. These results demonstrated that PET MP exposure induced male reproductive toxicity through oxidative stress and the p38 signaling pathway. This study provides new insights into the reproductive toxicity of MPs in males, as well as valuable references for public health protection strategies. Full article
Show Figures

Figure 1

Figure 1
<p>Body and testicular weight. (<b>a</b>) Body weight. (<b>b</b>) Testicular weight. <span class="html-italic">n</span> = 10 for all groups. Data are presented as the mean ± SEM.</p>
Full article ">Figure 2
<p>The pathological changes in testicular tissues determined via HE staining after PET MP exposure. (<b>a</b>) Control group; (<b>b</b>) 0.01 mg/d PET MP group; (<b>c</b>) 0.1 mg/d PET MP group; and (<b>d</b>) 1 mg/d PET MP group. The black arrow indicates that the spermatogenic tubule was empty. The red arrow indicates the disorder of the spermatogenic epithelium. The yellow arrow indicates the decrease in spermatogenic cells. Scale bar: 50 µm. <span class="html-italic">n</span> = 5 for all groups.</p>
Full article ">Figure 3
<p>The changes in sperm quality after PET MP exposure. (<b>a</b>) The number of sperm; (<b>b</b>) the rate of sperm viability; and (<b>c</b>) the rate of teratosperm. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. * indicates <span class="html-italic">p</span> &lt; 0.05 (compared with the control group). ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 4
<p>The effect of spermatogenic cell apoptosis via TUNEL staining after PET MP exposure. (<b>a</b>) Control group; (<b>b</b>) 0.01 mg/d PET MP group; (<b>c</b>) 0.1 mg/d PET MP group; (<b>d</b>) 1 mg/d PET MP group; and (<b>e</b>) the quantitative analysis of the apoptosis rate. The orange arrow indicates the apoptotic cells. Scale bar: 50 µm. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 5
<p>The expression levels of Caspase-3 and Caspase-9 after PET MP exposure. (<b>a</b>) The levels of Caspase-3; (<b>b</b>) the levels of Caspase-9. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 6
<p>GSH and MDA levels in testicular tissue after PET MP exposure. (<b>a</b>) GSH levels; (<b>b</b>) MDA levels. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 7
<p>The pathological changes in the testicular tissue of mice after NAC antagonism. (<b>a</b>) GSH levels; (<b>b</b>) MDA levels; (<b>c</b>) control group; (<b>d</b>) NAC-treated group; (<b>e</b>) 1 mg/d PET MP group; (<b>f</b>) NAC intervention group; (<b>g</b>) sperm count levels; (<b>h</b>) sperm viability rate; and (<b>i</b>) teratosperm rate. The black arrow indicates that the spermatogenic tubule was empty. The red arrow indicates a disorder of the spermatogenic epithelium. The yellow arrow indicates a decrease in spermatogenic cells. The purple arrow indicates that the spermatogenic tubules are partially restored. The brown arrow indicates that the spermatogenic cells gradually recovered. Scale bar: 50 µm. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 8
<p>The activation of the p38 MAPK signaling pathway after PET MP exposure. (<b>a</b>) The expression of p38 and p-p38 in testis; (<b>b</b>) the quantification of p38 and p-p38 protein levels. β-actin was considered as the loading control. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. * indicates <span class="html-italic">p</span> &lt; 0.05 (compared with the control group). ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the control group).</p>
Full article ">Figure 9
<p>The pathological changes in testicular tissue after SB203580 blockade. (<b>a</b>) The expression of p38 and p-p38 in testis; (<b>b</b>) the quantification of p38 and p-p38 protein levels. β-actin was considered as the loading control. (<b>c</b>) Control group; (<b>d</b>) SB203580-treated group; (<b>e</b>) 1 mg/d PET MP group; (<b>f</b>) SB203580 intervention group; (<b>g</b>) the levels of sperm count; (<b>h</b>) the rate of sperm viability; and (<b>i</b>) the rate of teratosperm. The black arrow indicates that the spermatogenic tubule is empty. The red arrow indicates a disorder of the spermatogenic epithelium. The yellow arrow indicates a decrease in spermatogenic cells. The purple arrow indicates that the spermatogenic tubules are partially restored. The brown arrow indicates that the spermatogenic cells gradually recovered. Scale bar: 50 µm. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the corresponding intervention group).</p>
Full article ">Figure 10
<p>The effect of apoptosis and apoptosis factors after SB203580 blockade. (<b>a</b>) Control group; (<b>b</b>) SB203580-treated group; (<b>c</b>) 1 mg/d PET MP group; (<b>d</b>) SB203580 intervention group; (<b>e</b>) the quantitative analysis of apoptosis rate; (<b>f</b>) Caspase-3 levels; and (<b>g</b>) Caspase-9 levels. The orange arrow indicates the apoptotic cells. Scale bar: 50 µm. <span class="html-italic">n</span> = 5 for all groups. Data are presented as the mean ± SEM. ** indicates <span class="html-italic">p</span> &lt; 0.01 (compared with the corresponding intervention group).</p>
Full article ">
15 pages, 6890 KiB  
Article
c-Jun N-terminal Kinase Supports Autophagy in Testicular Ischemia but Triggers Apoptosis in Ischemia-Reperfusion Injury
by Sarah R. Alotaibi, Waleed M. Renno and May Al-Maghrebi
Int. J. Mol. Sci. 2024, 25(19), 10446; https://doi.org/10.3390/ijms251910446 - 27 Sep 2024
Viewed by 806
Abstract
Oxidative stress triggered by testicular torsion and detorsion in young males could negatively impact future fertility. Using a rat animal model for testicular IRI (tIRI), we aim to study the induction of autophagy (ATG) during testicular ischemia and tIRI and the role of [...] Read more.
Oxidative stress triggered by testicular torsion and detorsion in young males could negatively impact future fertility. Using a rat animal model for testicular IRI (tIRI), we aim to study the induction of autophagy (ATG) during testicular ischemia and tIRI and the role of oxidative-stress-induced c-Jun N-terminal Kinase (JNK) as a cytoprotective mechanism. Sixty male Sprague-Dawley rats were divided into five groups: sham, ischemia only, ischemia+SP600125 (a JNK inhibitor), tIRI only, and tIRI+SP600125. The tIRI rats underwent an ischemic injury for 1 h followed by 4 h of reperfusion, while ischemic rats were subjected to 1 h of ischemia only without reperfusion. Testicular-ischemia-induced Beclin 1 and LC3B expression was associated with decreased p62/SQSTM1 expression, increased ATP and alkaline phosphatase (AP) activity, and slightly impaired spermatogenesis. SP600125 treatment improved p62 expression and reduced the levels of Beclin 1 and LC3B but did not affect ATP or AP levels. The tIRI-induced apoptosis lowered the expression of the three ATG proteins and AP activity, activated caspase 3, and caused spermatogenic arrest. SP600125-inhibited JNK during tIRI restored sham levels to all investigated parameters. This study emphasizes the regulatory role of JNK in balancing autophagy and apoptosis during testicular oxidative injuries. Full article
Show Figures

Figure 1

Figure 1
<p>JNK phosphorylation is associated with testicular ischemia and tIRI. (<b>a</b>) Representative Western blots (WB) of JNK and p-JNK (T183 and Y185) in ipsilateral testes of experimental groups: sham, ischemia, tIRI, ischemia+SP600125, and tIRI+SP600125. SP600125 is injected intraperitoneally 30 min prior to ischemia and tIRI at 15 mg/Kg. (<b>b</b>) WB band intensity of JNK and p-JNK normalized to GAPDH. (<b>c</b>) Ratio of p-JNK to JNK expression. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 2
<p>JNK modulates the gene expression of autophagy markers: Beclin 1, LC3B, and p62. (<b>a</b>) Representative Western blots and (<b>b</b>) bar graphs of Beclin 1, LC3B, and p62/SQSTM1 protein expression in all 5 experimental groups. (<b>c</b>) The relative mRNA expression of autophagy markers Becn1, Lc3b, and Sqstm1 was calculated using the 2<sup>−ΔΔCt</sup> method. The fold change in gene expression in ischemia-, tIRI-, and SP600125-treated groups was calculated relative to the sham group. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 3
<p>JNK modulates LC3B expression. LC3B was detected in seminiferous tubules using fluorescence confocal microscopy. Representative immunofluorescence (IF)-stained testicular tissue sections for (<b>a</b>) sham, (<b>b</b>) ischemia, (<b>c</b>) ischemia+SP600125, (<b>d</b>) tIRI, and (<b>e</b>) tIRI+SP600125 groups. (<b>f</b>) Quantification of the average IF intensity and statistical analysis. Images were taken at 40× magnification with a scale bar of 50 μm. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham.</p>
Full article ">Figure 4
<p>JNK modulates lysosomal alkaline phosphatase (AP) activity but not ATP levels. Biochemical colorimetric assay kits measured AP activity and ATP concentration in the 5 experimental groups. The data were analyzed using one-way analysis of variance (ANOVA) followed by the Holm–Sidak multiple comparisons test and presented as mean values ± SD (<span class="html-italic">n</span> = 6/group). * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">Figure 5
<p>JNK modulates caspase 3 activity. Caspase 3 activity was measured using a colorimetric assay, and the fold change in ischemia-, tIRI-, and SP600125-treated groups was calibrated to sham activity. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * tIRI compared to ischemia and # tIRI compared to sham.</p>
Full article ">Figure 6
<p>JNK modulates spermatogenesis activity. H&amp;E-stained testicular tissue sections for (<b>a</b>) sham, (<b>b</b>) ischemia, (<b>c</b>) ischemia+SP600125, (<b>d</b>) tIRI, and (<b>e</b>) tIRI+SP600125 groups. (<b>f</b>) Spermatogenesis analysis using the Johnsen score. Images were taken at 40× magnification with a scale bar of 200 μm. Data are presented as mean ± SD (<span class="html-italic">n</span> = 6/group), <span class="html-italic">p</span>-value &lt; 0.05. * Ischemia compared to sham and # tIRI compared to sham.</p>
Full article ">
15 pages, 1731 KiB  
Review
The Protective Role of L-Cysteine in the Regulation of Blood–Testis Barrier Functions—A Brief Review
by Jeffrey Justin Margret and Sushil K. Jain
Genes 2024, 15(9), 1201; https://doi.org/10.3390/genes15091201 - 12 Sep 2024
Cited by 1 | Viewed by 1183
Abstract
Blood–testis barrier (BTB) genes are crucial for the cellular mechanisms of spermatogenesis as they protect against detrimental cytotoxic agents, chemicals, and pathogens, thereby maintaining a sterile environment necessary for sperm development. BTB proteins predominantly consist of extensive tight and gap junctions formed between [...] Read more.
Blood–testis barrier (BTB) genes are crucial for the cellular mechanisms of spermatogenesis as they protect against detrimental cytotoxic agents, chemicals, and pathogens, thereby maintaining a sterile environment necessary for sperm development. BTB proteins predominantly consist of extensive tight and gap junctions formed between Sertoli cells. These junctions form a crucial immunological barrier restricting the intercellular movement of substances and molecules within the adluminal compartment. Epithelial tight junctions are complex membrane structures composed of various integral membrane proteins, including claudins, zonula occludens-1, and occludin. Inter-testicular cell junction proteins undergo a constant process of degradation and renewal. In addition, the downregulation of genes crucial to the development and preservation of cell junctions could disrupt the functionality of the BTB, potentially leading to male infertility. Oxidative stress and inflammation may contribute to disrupted spermatogenesis, resulting in male infertility. L-cysteine is a precursor to glutathione, a crucial antioxidant that helps mitigate damage and inflammation resulting from oxidative stress. Preclinical research indicates that L-cysteine may offer protective benefits against testicular injury and promote the expression of BTB genes. This review emphasizes various BTB genes essential for preserving its structural integrity and facilitating spermatogenesis and male fertility. Furthermore, it consolidates various research findings suggesting that L-cysteine may promote the expression of BTB-associated genes, thereby aiding in the maintenance of testicular functions. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Figure 1
<p>A graphical representation of the blood–testis barrier (BTB) located within the seminiferous epithelium.</p>
Full article ">Figure 2
<p>The beneficial effect of L-cysteine supplementation against the damages caused by various external and genetic factors affecting the integrity of BTB proteins leads to reduced permeability and gene expression. EDC, endocrine-disrupting compounds; ROS, reactive oxygen species.</p>
Full article ">
19 pages, 6423 KiB  
Article
Role of Anonychium africanum (Plantae, Fabaceae) in Metal Oxido-Inflammatory Response: Protection Evidence in Gonad of Male Albino Rat
by Harrison A. Ozoani, Orish Ebere Orisakwe, Costantino Parisi, Loredana Assisi, Anthonet N. Ezejiofor, Kenneth O. Okolo, Chinna N. Orish, Rubina Vangone, Emidio M. Sivieri and Giulia Guerriero
Antioxidants 2024, 13(9), 1028; https://doi.org/10.3390/antiox13091028 - 24 Aug 2024
Viewed by 1041
Abstract
Male fertility is strongly affected by the overexpression of free radicals induced by heavy metals. The aim of this study was to evaluate the potential antioxidant, anti-inflammatory, and gonado-protective effects of natural compounds. Biochemical and morphological assays were performed on male albino rats [...] Read more.
Male fertility is strongly affected by the overexpression of free radicals induced by heavy metals. The aim of this study was to evaluate the potential antioxidant, anti-inflammatory, and gonado-protective effects of natural compounds. Biochemical and morphological assays were performed on male albino rats divided into five groups: a control group (water only), a group orally exposed to a metal mixture of Pb-Cd-Hg-As alone and three groups co-administered the metal mixture and an aqueous extract of the Nigerian medicinal plant, Anonychium africanum (Prosopis africana, PA), at three different concentrations (500, 1000, and 1500 mg/kg) for 60 days. The metal mixture induced a significant rise in testicular weight, metal bioaccumulation, oxidative stress, and pro-inflammatory and apoptotic markers, while the semen analysis indicated a lower viability and a decrease in normal sperm count, and plasma reproductive hormones showed a significant variation. Parallel phytochemical investigations showed that PA has bioactive compounds like phlobatannins, flavonoids, polyphenols, tannins, saponins, steroids, and alkaloids, which are protective against oxidative injury in neural tissues. Indeed, the presence of PA co-administered with the metal mixture mitigated the toxic metals’ impact, which was determined by observing the oxido-inflammatory response via nuclear factor erythroid 2-related factor 2, thus boosting male reproductive health. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Experimental design: grouping, dose administration, and measured parameters.</p>
Full article ">Figure 2
<p>The impact of <span class="html-italic">Prosopis africana</span> (<span class="html-italic">PA</span>) on hormonal profile in plasma of male albino rats exposed to a heavy metal mixture (HMM) for 60 days. (<b>A</b>) the effect of <span class="html-italic">PA</span> on follicle-stimulating hormone (FSH). (<b>B</b>) The effect of <span class="html-italic">PA</span> on luteinizing hormone (LH). (<b>C</b>) The effect of <span class="html-italic">PA</span> on testosterone (T). (<b>D</b>) The effect of <span class="html-italic">PA</span> on prolactin (PRL). Values are mean ± SD, N = 7. Bars having the same letter notations (a, b, c, d) are not significantly different from each other (<span class="html-italic">p</span> ≥ 0.05).</p>
Full article ">Figure 3
<p>The concentration of heavy metal mixture (HMM) with and without <span class="html-italic">Prosopis africana</span> (<span class="html-italic">PA</span>) aqueous extracts on testicular tissue. (<b>A</b>) the concentration of lead (Pb) in testicular tissue exposed to HMM alone and the combination HMM and <span class="html-italic">PA</span>. (<b>B</b>) The concentration of cadmium (Cd) in testicular tissue exposed to HMM alone and the combination HMM and <span class="html-italic">PA</span>. (<b>C</b>) The concentration of arsenic (As) in testicular tissue exposed to HMM alone and the combination HMM and <span class="html-italic">PA</span>. Values are mean ± SD, N = 7. Bars having the same letter notations (a, b, c, d,) are not significantly different from each other (<span class="html-italic">p</span> ≥ 0.05).</p>
Full article ">Figure 4
<p>The impact of <span class="html-italic">Prosopis africana</span> (<span class="html-italic">PA</span>) on oxidative stress markers in male albino rats exposed for 60 days to a heavy metal mixture (HMM). (<b>A</b>) The effect of <span class="html-italic">PA</span> on SOD. (<b>B</b>) The effect of <span class="html-italic">PA</span> on CAT. (<b>C</b>) The effect of <span class="html-italic">PA</span> on GPx. (<b>D</b>) The effect of <span class="html-italic">PA</span> on GSH. (<b>E</b>) The effect of <span class="html-italic">PA</span> on MDA. (<b>F</b>) the effect of <span class="html-italic">PA</span> on NO. Values are mean ± SD, N = 7. Bars sharing the same letter notations (a, b, c, d, e) are not significantly different from each other (<span class="html-italic">p</span> ≥ 0.05).</p>
Full article ">Figure 5
<p>The impact of <span class="html-italic">Prosopis africana</span> (<span class="html-italic">PA</span>) on expression of pro-inflammatory factors and apoptotic and transcriptional factors in male albino rats exposed to a heavy metal mixture (HMM) for 60 days. (<b>A</b>) The effect of <span class="html-italic">PA</span> on interleukine-6 (IL-6). (<b>B</b>) The effect of <span class="html-italic">PA</span> on tumor necrotic factor alfa (TNF-α). (<b>C</b>) The effect of <span class="html-italic">PA</span> on caspase-3. (<b>D</b>) The effect of <span class="html-italic">PA</span> on nuclear factor kappa B (NF-κB). (<b>E</b>) The effect of <span class="html-italic">PA</span> on transcriptional factor Nrf2. Values are mean ± SD, N = 7. Bars sharing the same letter notations (a, b, c, d, e) are not significantly different from each other (<span class="html-italic">p</span> ≥ 0.05).</p>
Full article ">
13 pages, 10879 KiB  
Article
Rosmarinic Acid Attenuates Testicular Damage via Modulating Oxidative Stress and Apoptosis in Streptozotocin-Induced Diabetic Albino Mice
by Omar Al-khawaldeh, Zina M. Al-Alami, Osama Y. Althunibat, Tamer M. M. Abuamara, Afnan Mihdawi and Mohammad H. Abukhalil
Stresses 2024, 4(3), 505-517; https://doi.org/10.3390/stresses4030032 - 5 Aug 2024
Viewed by 1051
Abstract
Diabetes mellitus (DM) induces the production of reactive oxygen species, which may lead to cell injury and death. This study aimed to assess the effects of rosmarinic acid (RA) on testicular damage, oxidative stress, and apoptosis in streptozotocin (STZ)-induced diabetic albino mice. DM [...] Read more.
Diabetes mellitus (DM) induces the production of reactive oxygen species, which may lead to cell injury and death. This study aimed to assess the effects of rosmarinic acid (RA) on testicular damage, oxidative stress, and apoptosis in streptozotocin (STZ)-induced diabetic albino mice. DM in four- to six-week-old BALB/c male albino mice was induced via 50 mg/kg STZ, IP for 5 days. Twelve mice were randomly assigned into each of following groups: a control group, a diabetic (DM) group, RA5 mg/kg and RA15 mg/kg groups, and DM + RA5 mg/kg and DM + RA15 mg/kg groups. RA doses were intraperitoneally injected six times a week for seven weeks. Diabetes increased blood sugar and HbA1c levels and decreased all assessed sperm parameters. Testicular tissues of the diabetic mice showed increased lipid peroxidation, decreased reduced glutathione levels and catalase and superoxide dismutase activities, and increased apoptosis associated with histological abnormalities. Both RA doses had no effects on final body weight, blood sugar, and HbA1c in the diabetic mice. It is concluded that the administration of the potent antioxidant RA to diabetic mice improved the redox status in testicular tissues, protected them from diabetes-induced oxidative damage, and improved the quality of spermatozoa, mostly in a dose-dependent manner, which suggests a potential application value of RA in treating DM-related testicular injury and perhaps other complications. Full article
(This article belongs to the Section Animal and Human Stresses)
Show Figures

Figure 1

Figure 1
<p>Effects of RA on body weight, FBG levels, and HbA1c in diabetic mice. (<b>a</b>) Average mean body weight during the experimental period. (<b>b</b>) Average mean values of fasting glucose level before and during treatment. (<b>c</b>) HbA1c levels. All values are expressed as the mean ± SEM. The letters on the bars mean the following: a: significant difference when compared to the control group at <span class="html-italic">p</span> &lt; 0.05; b: significant difference when compared to the DM group at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Effects of RA on sperm parameters in diabetic mice: (<b>a</b>) Sperm count. (<b>b</b>) Percentage of motile sperms. (<b>c</b>) Percentage of viable sperm. (<b>d</b>) Percentage of sperm with normal morphology. All values are expressed as the mean ± SEM. The letters above the bars mean the following: a: significant difference when compared to the control group at <span class="html-italic">p</span> &lt; 0.05; b: significant difference when compared to the DM group at <span class="html-italic">p</span> &lt; 0.05; ab: significant difference when compared to the control and DM groups at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effects of RA on oxidative stress in the testis of diabetic mice: (<b>a</b>) TBARS levels. (<b>b</b>) Concentration of reduced glutathione (GSH). (<b>c</b>) SOD activity. (<b>d</b>) Catalase (CAT) absorbance. All values are expressed as the mean ± SEM. The letters above the bars mean the following: a: significant difference when compared to the control group at <span class="html-italic">p</span> &lt; 0.05; b: significant difference when compared to the DM group at <span class="html-italic">p</span> &lt; 0.05; ab: significant difference when compared to control and DM group at <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Photomicrographs obtained from the testes of mice from (<b>a</b>) the control group, (<b>b</b>) the DM group, (<b>c</b>) the RA5 mg/kg group, (<b>d</b>) the RA15 mg/kg group, (<b>e</b>) the DM + RA5 mg/kg group, and (<b>f</b>) the DM + RA15 mg/kg group (X400; H&amp;E); scale bar: 100 uM. Early spermatids (ES), fibroblasts (F), hyalinization in the lumen of some seminiferous tubules (H), interstitial cells of Leydig (LC), late spermatids (LS), myoid cells (M), primary spermatocytes (PS), pyknotic and necrotic spermatogenic cells (Pk), seminiferous tubules (ST), Sertoli cells (SC), spermatid giant cells (GC), spermatids (S), spermatogonia (SG), vacuoles (V). Sloughing germinal epithelium and irregular basement membrane (*), absence of spermatogenesis (red circle).</p>
Full article ">Figure 5
<p>Representative images of TUNEL-stained sections in the testes of mice: (<b>a</b>) Control group. (<b>b</b>) DM group. (<b>c</b>) RA5 mg/kg group. (<b>d</b>) RA15 mg/kg group. (<b>e</b>) DM + RA5 mg/kg group. (<b>f</b>) DM + RA15 mg/kg group. In these fluorescent microscopic images, the bright green color represents DNA-fragmented cells, and the blue color represents the DAPI stain, meaning intact DNA. Scale bar: 10 μm.</p>
Full article ">
12 pages, 3089 KiB  
Article
Rumen Microbiota Transplantation Alleviates Gossypol Diet-Induced Reproductive, Liver, and Intestinal Damage in Male Mice
by Chen Zhang, Wenguang Lu, Huiru Liu, Lingwei Shen, Mengfan Zhu, Tangtang Zhou, Ling Zhang, Dingfu Xiao and Lijuan Chen
Animals 2024, 14(15), 2206; https://doi.org/10.3390/ani14152206 - 30 Jul 2024
Cited by 1 | Viewed by 1078
Abstract
Ruminants exhibit stronger tolerance to gossypol, an anti-nutritional factor, compared to monogastric animals. We transplanted Hu sheep rumen microbiota into male mice to investigate the role of rumen microbiota in animal gossypol tolerance. Thirty specific-pathogen-free (SPF) male C57BL/6 mice were randomly divided into [...] Read more.
Ruminants exhibit stronger tolerance to gossypol, an anti-nutritional factor, compared to monogastric animals. We transplanted Hu sheep rumen microbiota into male mice to investigate the role of rumen microbiota in animal gossypol tolerance. Thirty specific-pathogen-free (SPF) male C57BL/6 mice were randomly divided into three groups: normal diet (CK group), gossypol diet (FG group), and rumen microbiota transplantation (FMT group, gossypol diet). The pathological changes in the liver and small intestine of the mice, the organ coefficient, and sperm parameters were analyzed. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in the blood and lactate dihydrogen-X (LDH-X) levels in the testicular tissue were also measured. The results showed that body weight, feed intake, sperm concentration, sperm motility, and LDH-X levels in the FMT group increased (p < 0.05) compared with the FG group, while the enzyme activities of ALT, AST, and AST/ALT decreased (p < 0.05). In the FMT group, the injury to liver cells was alleviated, the structure of the small intestine was intact, and the villus height and the ratio of villus height to crypt depth (V/C) were higher than those in the FG group (p < 0.05). And there were no differences in various organ coefficients and sperm deformity rates among the three groups (p > 0.05), but compared with the FG group, mice in the FMT group showed tendencies closer to those in the CK group. Rumen microbiota transplantation relieved the reproductive toxicity and liver damage induced by gossypol in male mice and improved the tolerance of recipient animals to gossypol. Additionally, rumen microbes improved the intestinal structural integrity of recipients. Full article
(This article belongs to the Special Issue Recent Advances in Probiotics Application on Animal Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Body weight and food intake of mice. Note: (<b>A</b>): Body weight of mice; (<b>B</b>): feed intake of mice. Values with different small-letter superscripts indicate a significant difference between groups at the same time (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Effects of FMT on organ coefficients in mice. Note: Value columns with the same or no small-letter superscripts mean no significant difference (<span class="html-italic">p</span> &gt; 0.05), while those with different small-letter superscripts indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>AST and ALT enzyme activities in serum. Note: (<b>A</b>): Alanine aminotransferase (ALT), (<b>B</b>): aspartate aminotransferase (AST), (<b>C</b>): AST/ALT ratio. Value columns with the same or no small-letter superscripts mean no significant difference (<span class="html-italic">p</span> &gt; 0.05), while those with different small-letter superscripts indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Effects of FMT on liver pathology (HE, ×400). Note: (<b>A</b>): CK group; (<b>B</b>): FG group; (<b>C</b>): FMT group.</p>
Full article ">Figure 5
<p>Effects of FMT on the reproductive function of male mice. Note: (<b>A</b>): Sperm concentration, (<b>B</b>): sperm motility, (<b>C</b>): sperm abnormality rate; (<b>D</b>): activity of LDH-X in the testis. Value columns with the same or no small-letter superscripts mean no significant difference (<span class="html-italic">p</span> &gt; 0.05), while those with different small-letter superscripts indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Morphology of the small intestine tissues of mice (HE, ×400). Note: (<b>A</b>): CK group; (<b>B</b>): FG group; (<b>C</b>): FMT group.</p>
Full article ">Figure 7
<p>Parameters of small intestine tissue in mice. Note: (<b>A</b>): Villus height, (<b>B</b>): crypt depth, (<b>C</b>): V/C rate. Value columns with the same or no small-letter superscripts mean no significant difference (<span class="html-italic">p</span> &gt; 0.05), while those with different small-letter superscripts indicate a significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
15 pages, 10668 KiB  
Article
Ganoderma lucidum Polysaccharide Peptide Alleviates Cyclophosphamide-Induced Male Reproductive Injury by Reducing Oxidative Stress and Apoptosis
by Hang Zhang, Nannan Li, Yukun Zhang, Yue Xu, Feng Lu, Dongmei Lin, Shuqian Lin, Min Li and Baoxue Yang
Biomedicines 2024, 12(8), 1632; https://doi.org/10.3390/biomedicines12081632 - 23 Jul 2024
Viewed by 1348
Abstract
Chemotherapy is an important factor leading to male infertility. It is crucial to discover safe and effective treatments to prevent male reproductive injury caused by chemotherapy. The Ganoderma lucidum polysaccharide peptide (GLPP) has multiple pharmacological activities. The purpose of this study was to [...] Read more.
Chemotherapy is an important factor leading to male infertility. It is crucial to discover safe and effective treatments to prevent male reproductive injury caused by chemotherapy. The Ganoderma lucidum polysaccharide peptide (GLPP) has multiple pharmacological activities. The purpose of this study was to determine whether GLPP could protect the male sperm production from chemotherapeutic injury using a mouse model, with testicular damage induced by cyclophosphamide (CP). CP (50 mg/kg/day) was injected intraperitoneally into male ICR mice gavaged with different doses of GLPP at certain spermatogenic stages. The experimental results showed that GLPP alleviated the CP-induced reduction in reproductive organ coefficients and sperm parameters and reduced the morphological damage of testicular tissues in a dose-dependent manner. GLPP significantly improved the reproductive index, sperm-related parameters, sex hormone levels, and histological testis architecture at different spermatogenic stages. Furthermore, GLPP significantly increased superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), Nrf2, and HO-1, and decreased malondialdehyde (MDA) and Keap-1 in the testicular tissue, indicating reduced oxidative stress. In addition, GLPP limited CP-induced apoptosis via a reduction in Bax expression and increase in Bcl-2 expression. This study suggests that GLPP plays a protective role in spermatogenesis by reducing chemotherapeutic injury and might be developed into drug for male patients receiving chemotherapy. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The schedule of the animal experiments.</p>
Full article ">Figure 2
<p>Effect of GLPP on CP-induced male reproductive injury in mice. (<b>A</b>) Testicular index. (<b>B</b>) Epididymal index. (<b>C</b>) Sperm count. (<b>D</b>) The representative photographs of sperm morphology from epididymis. The yellow arrows indicate the sperm with abnormal morphology (<b>E</b>) Sperm abnormality. (<b>F</b>) Sperm motility. Data are presented as mean ± SEM (n = 6). ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. Ctr; * <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 vs. CP; ns, no significance.</p>
Full article ">Figure 3
<p>Histopathology of testis. (<b>A</b>) Representative images of testicular H&amp;E staining. (<b>B</b>) Johnsen score of the seminiferous tubules. Data are presented as mean ± SEM (n = 6). ### <span class="html-italic">p</span> &lt; 0.001 vs. Ctr; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. CP; ns, no significance.</p>
Full article ">Figure 4
<p>Organ indexes and sperm parameters. (<b>A</b>) Testicular index. (<b>B</b>) Epididymal index. (<b>C</b>) Sperm count. (<b>D</b>) Sperm abnormality. (<b>E</b>–<b>H</b>) Sperm kinematic parameters. (<b>I</b>) Sperm motility. Data are presented as mean ± SEM (n = 6). # <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 vs. Ctr; * <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 vs. CP; ns, no significance.</p>
Full article ">Figure 5
<p>Tissue morphology of the testis. (<b>A</b>) Representative images of testicular H&amp;E staining. (<b>B</b>) Johnsen’s score of the seminiferous tubules. Data were presented as mean ± SEM (n = 6). ### <span class="html-italic">p</span> &lt; 0.001 vs. Ctr; *** <span class="html-italic">p</span> &lt; 0.001 vs. CP; ns, no significance.</p>
Full article ">Figure 6
<p>GSH, SOD, CAT activity and MDA content in the testicular tissue. (<b>A</b>) GSH activity. (<b>B</b>) SOD activity. (<b>C</b>) CAT activity. (<b>D</b>) MDA content. Data were presented as mean ± SEM (n = 6). ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. Ctr; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. CP; ns, no significance.</p>
Full article ">Figure 7
<p>Testosterone and follicle-stimulating hormone (FSH) concentrations in mouse serum. (<b>A</b>) Testosterone concentration. (<b>B</b>) FSH concentration. Data were presented as mean ± SEM (n = 3). ## <span class="html-italic">p</span> &lt; 0.01; * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. CP; ns, no significance.</p>
Full article ">Figure 8
<p>Keap-1, Nrf2, and HO-1 protein expressions in testicular tissue. (<b>A</b>) Representative Western blots of Keap-1, Nrf2, HO-1 and β-actin. Relative levels of Keap1 (<b>B</b>), Nrf2 (<b>C</b>) and HO-1 (<b>D</b>) quantified by the band intensity. Data were presented as mean ± SEM (n = 4). # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. Ctr; * <span class="html-italic">p</span> &lt; 0.05 vs. CP; ns, no significance.</p>
Full article ">Figure 9
<p>Protein expressions involved in Bax/Bcl-2 signaling pathway in the testicular tissue. (<b>A</b>) Representative Western blots of Bax, Bcl-2, and β-actin. The relative protein expression levels of Bax (<b>B</b>), Bcl-2 (<b>C</b>) and Bax/Bcl-2 ratio (<b>D</b>). Data are presented as mean ± SEM (n = 4). # <span class="html-italic">p</span> &lt; 0.05 vs. Ctr; * <span class="html-italic">p</span> &lt; 0.05 vs. CP; ns, no significance.</p>
Full article ">
26 pages, 11705 KiB  
Article
Targeting JAK2/STAT3, NLRP3/Caspase-1, and PK2/PKR2 Pathways with Arbutin Ameliorates Lead Acetate-Induced Testicular Injury in Rats
by Hany H. Arab, Shuruq E. Alsufyani, Ahmed M. Ashour, Amany M. Gad, Alzahraa A. Elhemiely, Mohamed H. A. Gadelmawla, Marwa Ahmed Mahmoud and Ali Khames
Pharmaceuticals 2024, 17(7), 909; https://doi.org/10.3390/ph17070909 - 8 Jul 2024
Viewed by 1274
Abstract
The reproductive system of males is adversely impacted by lead (Pb), a toxic heavy metal. The present study examined arbutin, a promising hydroquinone glycoside, for its potential ameliorative impact against Pb-induced testicular impairment in rats. The testicular injury was induced by the intraperitoneal [...] Read more.
The reproductive system of males is adversely impacted by lead (Pb), a toxic heavy metal. The present study examined arbutin, a promising hydroquinone glycoside, for its potential ameliorative impact against Pb-induced testicular impairment in rats. The testicular injury was induced by the intraperitoneal administration of Pb acetate (20 mg/kg/day) for 10 consecutive days. Thirty-six rats were divided into six experimental groups (n = 6 per group): control, control treated with oral arbutin (250 mg/kg), control treated with intraperitoneal arbutin (75 mg/kg), untreated Pb, Pb treated with oral arbutin, and Pb treated with intraperitoneal arbutin. The treatments were administered daily for 10 days. Arbutin was administered by the oral and intraperitoneal routes to compare the efficacy of both routes in mitigating Pb acetate-induced testicular dysfunction. The current data revealed that both oral and intraperitoneal administration of arbutin significantly enhanced serum testosterone and sperm count/motility, indicating the amelioration of testicular dysfunction. In tandem, both routes lowered testicular histopathological aberrations and Johnsen’s damage scores. These favorable outcomes were driven by dampening testicular oxidative stress, evidenced by lowered lipid peroxidation and increased glutathione and catalase antioxidants. Moreover, arbutin lowered testicular p-JAK2 and p-STAT3 levels, confirming the inhibition of the JAK2/STAT3 pro-inflammatory pathway. In tandem, arbutin suppressed the testicular NLRP3/caspase-1/NF-B axis and augmented the cytoprotective PK2/PKR2 pathway. Notably, intraperitoneal arbutin at a lower dose prompted a more pronounced mitigation of Pb-induced testicular dysfunction compared to oral administration. In conclusion, arbutin ameliorates Pb-evoked testicular damage by stimulating testicular antioxidants and the PK2/PKR2 pathway and inhibiting the JAK2/STAT3 and NLRP3/caspase-1 pro-inflammatory pathways. Hence, arbutin may be used as an adjunct agent for mitigating Pb-induced testicular impairment. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Arbutin’s chemical structure.</p>
Full article ">Figure 2
<p>Ameliorative effect of arbutin (ARB), administered either orally or intraperitoneally, on sperm characteristics in lead (Pb)-evoked testicular damage in rats. ARB supplementation augmented sperm count (<b>A</b>) and sperm motility (<b>B</b>) while lowering sperm abnormalities (<b>C</b>) in the treated animals. Data represented as mean ± SEM (<span class="html-italic">n</span> = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 2 Cont.
<p>Ameliorative effect of arbutin (ARB), administered either orally or intraperitoneally, on sperm characteristics in lead (Pb)-evoked testicular damage in rats. ARB supplementation augmented sperm count (<b>A</b>) and sperm motility (<b>B</b>) while lowering sperm abnormalities (<b>C</b>) in the treated animals. Data represented as mean ± SEM (<span class="html-italic">n</span> = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 3
<p>Ameliorative effect of arbutin (ARB) given orally or intraperitoneally on serum sex hormones in lead (Pb)-evoked testicular damage in rats. ARB boosted serum testosterone (<b>A</b>) and counteracted the increased serum LH (<b>B</b>) and FSH (<b>C</b>) levels. Moreover, ARB augmented testicular acid phosphatase activity in animals (<b>D</b>). Data represented as mean ± SEM (<span class="html-italic">n</span> = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 or <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 4
<p>Photomicrographs of testicular tissue stained with hematoxylin and eosin (H-E) (scale bar: 20 µm). The photomicrographs are representative of n = 6 tissues. (<b>A</b>) In control animals, intact testicular architecture was observed with typical seminiferous tubules (STs) showing sperm (S), germinal cells (G), spermatocyte (thin arrow), spermatid (wavy arrow), spermatogonia (bifid arrow), and interstitial cells. Additionally, the interstitial space contained clusters of Leydig interstitial cells (<b>B</b>,<b>C</b>) Lead (Pb) acetate-challenged animals showed seminiferous tubule lumen lacking spermatozoa (L), a loss of germinal epithelium series (Line), and vacuolation (arrowhead). Intact testicular architecture with typical seminiferous tubules and intact interstitial cells (star) was shown in the control groups treated with oral (<b>D</b>) or intraperitoneal arbutin (ARB; (<b>E</b>)). In the groups co-treated with Pb and ARB via oral (<b>F</b>) or intraperitoneal injection (<b>G</b>), active seminiferous tubules and intact interstitial cells (star)were detected, implying attenuated testicular histopathological damage. (<b>H</b>) Spermatogenesis was scored according to Johnsen’s scoring scale. To present the scores, the medians and interquartile ranges are displayed (<span class="html-italic">n</span> = 6). The scores support the competence of oral and intraperitoneal administration of ARB to rescue the spermatogenesis process. Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4 Cont.
<p>Photomicrographs of testicular tissue stained with hematoxylin and eosin (H-E) (scale bar: 20 µm). The photomicrographs are representative of n = 6 tissues. (<b>A</b>) In control animals, intact testicular architecture was observed with typical seminiferous tubules (STs) showing sperm (S), germinal cells (G), spermatocyte (thin arrow), spermatid (wavy arrow), spermatogonia (bifid arrow), and interstitial cells. Additionally, the interstitial space contained clusters of Leydig interstitial cells (<b>B</b>,<b>C</b>) Lead (Pb) acetate-challenged animals showed seminiferous tubule lumen lacking spermatozoa (L), a loss of germinal epithelium series (Line), and vacuolation (arrowhead). Intact testicular architecture with typical seminiferous tubules and intact interstitial cells (star) was shown in the control groups treated with oral (<b>D</b>) or intraperitoneal arbutin (ARB; (<b>E</b>)). In the groups co-treated with Pb and ARB via oral (<b>F</b>) or intraperitoneal injection (<b>G</b>), active seminiferous tubules and intact interstitial cells (star)were detected, implying attenuated testicular histopathological damage. (<b>H</b>) Spermatogenesis was scored according to Johnsen’s scoring scale. To present the scores, the medians and interquartile ranges are displayed (<span class="html-italic">n</span> = 6). The scores support the competence of oral and intraperitoneal administration of ARB to rescue the spermatogenesis process. Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Mitigating effects of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular prooxidant/antioxidant events in lead (Pb)-evoked testicular damage in rats. ARB dampened the prooxidant lipid peroxides (<b>A</b>) and augmented reduced glutathione (GSH; (<b>B</b>)) and catalase activity (<b>C</b>) in the testicular tissues of animals. Data represented as mean ± SEM (n = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 5 Cont.
<p>Mitigating effects of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular prooxidant/antioxidant events in lead (Pb)-evoked testicular damage in rats. ARB dampened the prooxidant lipid peroxides (<b>A</b>) and augmented reduced glutathione (GSH; (<b>B</b>)) and catalase activity (<b>C</b>) in the testicular tissues of animals. Data represented as mean ± SEM (n = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 6
<p>Modulatory effect of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular inflammatory signals in lead (Pb)-evoked testicular damage in rats. (<b>A</b>) Immunoblot analysis demonstrated the protein expression of p-JAK2, NLRP3, and PK2. To verify that total protein lysates were equally loaded, beta-actin was utilized as the loading control. (<b>B</b>) p-JAK2 quantification. (<b>C</b>) NLRP3 quantification. (<b>D</b>) PK2 quantification. In the Western blotting experiment, 1.0 was set as the control value. Data for the values in (<b>B</b>–<b>D</b>) are shown as mean ± SEM (from 3 independent experiments). (<b>E</b>) ARB lowers p-STAT3 in the testicular tissues of rats challenged with Pb. Data representation as mean ± SEM (n = 6). Compared to the control group, significance was described by ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group: significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 6 Cont.
<p>Modulatory effect of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular inflammatory signals in lead (Pb)-evoked testicular damage in rats. (<b>A</b>) Immunoblot analysis demonstrated the protein expression of p-JAK2, NLRP3, and PK2. To verify that total protein lysates were equally loaded, beta-actin was utilized as the loading control. (<b>B</b>) p-JAK2 quantification. (<b>C</b>) NLRP3 quantification. (<b>D</b>) PK2 quantification. In the Western blotting experiment, 1.0 was set as the control value. Data for the values in (<b>B</b>–<b>D</b>) are shown as mean ± SEM (from 3 independent experiments). (<b>E</b>) ARB lowers p-STAT3 in the testicular tissues of rats challenged with Pb. Data representation as mean ± SEM (n = 6). Compared to the control group, significance was described by ** <span class="html-italic">p</span> &lt; 0.01, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group: significance was described by <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 7
<p>Modulatory effect of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular content of ASC and caspase 1 in lead (Pb)-evoked testicular damage in rats. (<b>A</b>) ARB lowered ASC in the testicular tissues of rats challenged with Pb. (<b>B</b>) Testicular tissues of rats challenged with Pb exhibited a reduction in caspase 1 content following the administration of ARB. Data represented as mean ± SEM (n = 6). Compared to the control group, significance was described by ** <span class="html-italic">p</span> &lt; 0.0, or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 8
<p>Ameliorative effect of arbutin (ARB), administered either orally or via intraperitoneal injection, on testicular protein expression of activated NF-κBp65 in lead (Pb)-evoked testicular damage in rats. The photomicrographs are representative of n = 6 tissues. (<b>A</b>) Photomicrographs of testicular tissues of all experimental groups. The immunoreactivity of NF-κBp65 was visualized in the tissues as a brown color developed by DAB chromogen (50-µm scale bar; positive staining is indicated by red arrows). (<b>B</b>) Quantification of NF-κBp65 expression in the testicular tissues. Data represented as mean ± SEM (n = 6). Compared to the control group, significance was described by ** <span class="html-italic">p</span> &lt; 0.01 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 9
<p>Ameliorative effect of arbutin (ARB), administered either orally or intraperitoneally, on testicular protein expression of caspase 3 and PKR2 in lead (Pb)-evoked testicular damage in rats. The photomicrographs are representative of n = 6 tissues. (<b>A</b>) Photomicrographs of testicular tissues of all experimental groups. The immunoreactivity of caspase 3 was observed in the tissues as a brown color developed by DAB chromogen (50-µm scale bar; positive staining is indicated by red arrows). (<b>B</b>) Quantification of caspase 3 expression in the testicular tissues. (<b>C</b>) ARB boosted the expression of PKR2 in the testicular tissues of rats challenged with Pb. Data represented as mean ± SEM (n = 6). Compared to the control group, significance was described by * <span class="html-italic">p</span> &lt; 0.05 or *** <span class="html-italic">p</span> &lt; 0.001. Compared to the untreated Pb group, significance was described by <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 or <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (as determined by Bonferroni’s test and one-way ANOVA).</p>
Full article ">Figure 10
<p>A summary of the current study’s outcomes and the molecular events through which arbutin mitigated Pb-triggered testicular dysfunction. The observed molecular changes of arbutin included the following. (A) Dampening the testicular inflammatory events by inhibiting the JAK2/STAT3 pro-inflammatory pathway in Pb-intoxicated rats. (B) Suppression of the inflammation-associated NLRP3/ASC/caspase-1 pathway in the testicular tissues. (C) Stimulation of the testicular cytoprotective PK2/PKR2 pathway, promoting the protection and survival of testicular germ cells. (D) Interference with the testicular prooxidant response and augmentation of antioxidant signals. In the figure, a solid arrow depicts activation, while a blunt arrow indicates inhibition.</p>
Full article ">Figure 11
<p>Summary of the experimental design. ARB, arbutin; Pb, lead.</p>
Full article ">
21 pages, 29438 KiB  
Article
Arginine Biosynthesis Mediates Wulingzhi Extract Resistance to Busulfan-Induced Male Reproductive Toxicity
by Zifang Wu, Yuxuan Ma, Shaoxian Chen, Yuyan Liu, Xianglin Liu, Heran Cao, Tianqi Jin, Long Li, Mengqi Huang, Fangxia Yang and Wuzi Dong
Int. J. Mol. Sci. 2024, 25(12), 6320; https://doi.org/10.3390/ijms25126320 - 7 Jun 2024
Viewed by 1130
Abstract
Busulfan, an indispensable medicine in cancer treatment, can cause serious reproductive system damage to males as a side effect of its otherwise excellent therapeutic results. Its widespread use has also caused its accumulation in the environment and subsequent ecotoxicology effects. As a Chinese [...] Read more.
Busulfan, an indispensable medicine in cancer treatment, can cause serious reproductive system damage to males as a side effect of its otherwise excellent therapeutic results. Its widespread use has also caused its accumulation in the environment and subsequent ecotoxicology effects. As a Chinese medicine, Wulingzhi (WLZ) has the effects of promoting blood circulation and improving female reproductive function. However, the potential effects of WLZ in male reproduction and in counteracting busulfan-induced testis damage, as well as its probable mechanisms, are still ambiguous. In this study, busulfan was introduced in a mouse model to evaluate its production of the testicular damage. The components of different WLZ extracts were compared using an untargeted metabolome to select extracts with greater efficacy, which were further confirmed in vivo. Here, we demonstrate abnormal spermatogenesis and low sperm quality in busulfan-injured testes. The WLZ extracts showed a strong potential to rehabilitate the male reproductive system; this effect was more prominent in room-temperature extracts. Additionally, both water and ethanol WLZ extracts at room temperature alleviated various busulfan-induced adverse effects. In particular, WLZ recovered spermatogenesis, re-activated arginine biosynthesis, and alleviated the increased oxidative stress and inflammation in the testis, ultimately reversing the busulfan-induced testicular injury. Collectively, these results suggest a promising approach to protecting the male reproductive system from busulfan-induced adverse side effects, as well as those of other similar anti-cancer drugs. Full article
(This article belongs to the Special Issue Molecular Research of Reproductive Toxicity)
Show Figures

Figure 1

Figure 1
<p>Effect of busulfan on testes of mice. (<b>A</b>) Schematic diagram of busulfan treatment during the test period. (<b>B</b>) Bright-field diagram of testicular size in control and busulfan injection groups. (<b>C</b>) Average testis weight. (<b>D</b>) Ratio of testis weight/body weight. (<b>E</b>) Average epididymis weight. (<b>F</b>) Ratio of epididymis weight/body weight. (<b>G</b>) Representative images of H&amp;E staining in testis sections, with the 2nd row showing magnifications of the boxed regions in the 1st row; scale bar = 100 μm (1st row) and 10 μm (2nd row). (<b>H</b>) The percentage of normal spermatogenesis tubules in testicular samples. (<b>I</b>) Sperm concentration in epididymides. (<b>J</b>,<b>K</b>) Motility (<b>J</b>) and progressive motility (<b>K</b>) of sperm were assessed by computer-assisted semen analysis. <span class="html-italic">n</span> = 6 for each group. All data are presented as means ± SEM. Statistical significance was determined by the unpaired Student’s <span class="html-italic">t</span>-test. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>The composition changes in different WLZ extracts. (<b>A</b>) The untargeted metabolome profiles generated on different WLZ extract samples using an LC-MS (Liquid Chromatograph–Mass Spectrometer). WLZ (w) RT, the WLZ water extract at room temperature; WLZ (e) RT, the WLZ ethanol extract at room temperature; WLZ (w) H, the WLZ water extract at 85 °C; WLZ (e) H, the WLZ ethanol extract at 85 °C. (<b>B</b>–<b>E</b>) The components found in different WLZ extracts. (<b>F</b>,<b>G</b>) The Venn diagrams generated to describe the common and unique metabolites in the WLZ water extract (<b>F</b>) and WLZ ethanol extract (<b>G</b>) at different temperatures. (<b>H</b>–<b>K</b>) The components of the unique metabolites in WLZ (w) RT (<b>H</b>), WLZ (e) RT (<b>I</b>), WLZ (w) H (<b>J</b>), and WLZ (e) H (<b>K</b>). <span class="html-italic">n</span> = 3 for each group.</p>
Full article ">Figure 3
<p>The metabolome changes in the WLZ water extract and WLZ ethanol extract at different temperatures. (<b>A</b>–<b>E</b>) The metabolomic changes in WLZ (w) RT and WLZ (w) H; (<b>A</b>) the 3D principal component analysis (PCA) score plot; (<b>B</b>) volcano plot showing the changes in metabolites; (<b>C</b>) heatmap of the top 50 differential metabolites; (<b>D</b>,<b>E</b>) the KEGG enrichment analysis of differential metabolites in WLZ (w) RT and WLZ (w) H. (<b>F</b>–<b>J</b>) The metabolomics changes in WLZ (e) RT and WLZ (e) H; (<b>F</b>) the 3D principal component analysis (PCA) score plot; (<b>G</b>) volcano plot showing the changes in metabolites; (<b>H</b>) heatmap of the top 50 differential metabolites; (<b>I</b>,<b>J</b>) the KEGG enrichment analysis of differential metabolites in WLZ (e) RT and WLZ (e) H. (<b>K</b>,<b>L</b>) The bar plot based on the differential metabolites which were enriched in the reproduction-related classes. <span class="html-italic">n</span> = 3 for each group.</p>
Full article ">Figure 4
<p>The metabolome changes in WLZ water and ethanol extracts at room temperature. (<b>A</b>–<b>C</b>) The Venn diagram was generated to describe the common and unique metabolites in the WLZ water extract and WLZ ethanol extract at room temperature, and the components of the unique metabolites. (<b>D</b>) The 3D principal component analysis (PCA) score plot. (<b>E</b>) Volcano plot showing the changes in metabolites. (<b>F</b>) Heatmap of the top 50 differential metabolites. (<b>G</b>,<b>H</b>) The pathway enrichment analysis of differential metabolites in WLZ (w) RT and WLZ (e) RT. The darker the color and larger the shape of the circle, the stronger the pathway influence. (<b>I</b>,<b>J</b>) The KEGG enrichment analysis of differential metabolites in WLZ (w) RT and WLZ (e) RT. <span class="html-italic">n</span> = 3 for each group.</p>
Full article ">Figure 5
<p>Effects of different WLZ extracts on testis injury caused by busulfan. (<b>A</b>) Schematic diagram of experiment. (<b>B</b>) Bright-field diagram of testicular size. (<b>C</b>) Average testis weight. (<b>D</b>) Ratio of testis weight/body weight. (<b>E</b>) Representative images of H&amp;E staining in testis sections, with the 2nd row showing magnifications of the boxed regions in the 1st row; scale bar = 100 μm (1st row) and 10 μm (2nd row). (<b>F</b>) The percentage of normal spermatogenesis tubules in testicular samples. (<b>G</b>–<b>I</b>) The tubular diameter (<b>G</b>), epithelium height (<b>H</b>), and lumen width (<b>I</b>) of the testis. <span class="html-italic">n</span> = 6 for each group. All data are presented as means ± SEM. Statistical analysis was conducted using one-way ANOVA followed by Sidak’s multiple comparisons test. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Changes in sperm parameters and marker genes involving spermatogenesis after WLZ treatment. (<b>A</b>) Statistical analysis of sperm concentration. (<b>B</b>,<b>C</b>) Motility and progressive motility of sperm. (<b>D</b>–<b>R</b>) Real-time PCR analysis of the mRNA expression of late-spermatogenic-event-related markers (<b>D</b>–<b>H</b>), spermatogonia-related markers (<b>I</b>–<b>M</b>), spermatocyte-related markers (<b>N</b>,<b>O</b>), and spermatid-related markers (<b>P</b>,<b>R</b>). <span class="html-italic">n</span> = 6 for each group. All data are presented as means ± SEM. Statistical analysis was performed using one-way ANOVA followed by Sidak’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>WLZ regulated arginine biosynthesis to affect the oxidative stress and inflammation in the testes. (<b>A</b>) Real-time PCR analysis of the mRNA expression of arginine biosynthesis-related markers. (<b>B</b>) Gene expression in arginine biosynthesis pathway. Green represents down-regulated genes, while red represents up-regulated genes in the Bus group. (<b>C</b>,<b>D</b>) Real-time PCR analysis of the mRNA expression of oxidative stress-related markers (<b>C</b>) and inflammation-related markers (<b>D</b>). (<b>E</b>) Sperman’s correlation between testis-related indicators and arginine-related indicators. (<b>F</b>) The correlation network of significant markers in (<b>E</b>). <span class="html-italic">n</span> = 6 for each group. All data are presented as means ± SEM. Statistical analysis was conducted using one-way ANOVA followed by Sidak’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Schematic diagram for the hypothetical molecular mechanism of arginine biosynthesis-mediated WLZ resistance to busulfan-induced testicular dysfunction. Busulfan exposure led to impaired testicular function, mainly manifesting as oxidative stress and inflammation of the testes, abnormal spermatogenesis, and decreased sperm parameters, while WLZ supplementation induced arginine biosynthesis, which then relieved the increased oxidative stress and inflammation in the testes and finally rescued the spermatogenesis and testis function injured by busulfan.</p>
Full article ">
23 pages, 4288 KiB  
Article
Annona squamosa Fruit Extract Ameliorates Lead Acetate-Induced Testicular Injury by Modulating JAK-1/STAT-3/SOCS-1 Signaling in Male Rats
by Maysa A. Mobasher, Alaa Muqbil Alsirhani, Maha Abdullah Alwaili, Fadi Baakdah, Thamir M Eid, Fahad A. Alshanbari, Reem Yahya Alzahri, Sahar Abdulrahman Alkhodair and Karim Samy El-Said
Int. J. Mol. Sci. 2024, 25(10), 5562; https://doi.org/10.3390/ijms25105562 - 20 May 2024
Cited by 5 | Viewed by 1572
Abstract
Lead (Pb) is a common pollutant that is not biodegradable and gravely endangers the environment and human health. Annona squamosa fruit has a wide range of medicinal uses owing to its phytochemical constituents. This study evaluated the effect of treatment with A. squamosa [...] Read more.
Lead (Pb) is a common pollutant that is not biodegradable and gravely endangers the environment and human health. Annona squamosa fruit has a wide range of medicinal uses owing to its phytochemical constituents. This study evaluated the effect of treatment with A. squamosa fruit extract (ASFE) on testicular toxicity induced in male rats by lead acetate. The metal-chelating capacity and phytochemical composition of ASFE were determined. The LD50 of ASFE was evaluated by probit analysis. Molecular docking simulations were performed using Auto Dock Vina. Forty male Sprague Dawley rats were equally divided into the following groups: Gp1, a negative control group; Gp2, given ASFE (350 mg/kg body weight (b. wt.)) (1/10 of LD50); Gp3, given lead acetate (PbAc) solution (100 mg/kg b. wt.); and Gp4, given PbAc as in Gp3 and ASFE as in Gp2. All treatments were given by oro-gastric intubation daily for 30 days. Body weight changes, spermatological parameters, reproductive hormone levels, oxidative stress parameters, and inflammatory biomarkers were evaluated, and molecular and histopathological investigations were performed. The results showed that ASFE had promising metal-chelating activity and phytochemical composition. The LD50 of ASFE was 3500 mg/kg b. wt. The docking analysis showed that quercetin demonstrated a high binding affinity for JAK-1 and STAT-3 proteins, and this could make it a more promising candidate for targeting the JAK-1/STAT-3 pathway than others. The rats given lead acetate had defective testicular tissues, with altered molecular, biochemical, and histological features, as well as impaired spermatological characteristics. Treatment with ASFE led to a significant mitigation of these dysfunctions and modulated the JAK-1/STAT-3/SOCS-1 axis in the rats. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>The most abundant phytochemicals in ASFE.</p>
Full article ">Figure 2
<p>Best compounds’ interactions with JAK-1, STAT-3, and SOCS-1 proteins (2D) and (3D). (<b>A</b>) Quercetin derivative with JAK-1 (−9.1). The conventional hydrogen bond (GLU83-2.71A), conventional hydrogen bond (ARG108-2.47A), conventional hydrogen bond (TYR412-2.86A), conventional hydrogen bond (MET109-2.68A), conventional hydrogen bond (TYR112-2.24A), conventional hydrogen bond (TYR281-2.42A), pi–pi T-shaped (TY) TYR281-5.50A), pi–anion (GLU284-4.96A), carbon–hydrogen bond (PRO172-3.77A). (<b>B</b>) β-sitosterol with SOCS-1 (−7.1). Pi–alkyl (PHE56-4.80, -5.21A), pi–sigma (THR65-3.97A). (<b>C</b>) Quercetin derivative with STAT-3 (−7.9). Conventional hydrogen bond (CYS712-3.41A), conventional hydrogen bond (GLU638-3.00A), carbon–hydrogen bond (VAL637-3.45A), pi–alkyl (PRO728-4.54, -5.40A), pi–sigma (THR714-3.60A).</p>
Full article ">Figure 3
<p>The median lethal dose (LD<sub>50</sub>) of ASFE.</p>
Full article ">Figure 4
<p>(<b>A</b>) The initial and final body weights and the percentages of the body weight changes. (<b>B</b>) The epididymis and testis weights in the different groups under study. I.B.W: initial body weight, F.B.W: final body weight, ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: lead acetate. Data are expressed as mean ± S.D. (<span class="html-italic">n</span> = 10). Means that do not share a letter showed significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Effect of administrated PbAc plus ASFE on lead residues in the testis of male rats. ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: Lead acetate. Data are expressed as mean ± S.D. (<span class="html-italic">n</span> = 10). Means that do not share a letter showed significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>(<b>A</b>) Luteinizing hormone (LH), (<b>B</b>) follicular stimulating hormone (FSH), and (<b>C</b>) testosterone. ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: lead acetate. Data are expressed as mean ± S.D. (<span class="html-italic">n</span> = 10). Means that do not share a letter showed significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>Effect of ASFE on the relative mRNA expression of <span class="html-italic">JAK-1</span> (<b>A</b>), STAT-3 (<b>B</b>), and <span class="html-italic">SOCS-1</span> (<b>C</b>) genes in lead acetate-intoxicated rats. ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: lead acetate, <span class="html-italic">JAK-1</span>: Janus kinase-1; <span class="html-italic">STAT-3</span>: signal transducer and activator of transcription-3; <span class="html-italic">SOCS-1</span>: suppressor of cytokine signaling-1. Data are expressed as mean ± S.D. (<span class="html-italic">n</span> = 10). Means that do not share a letter showed significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 8
<p>Effect of ASFE on the relative mRNA expression of <span class="html-italic">IL-6</span> (<b>A</b>), <span class="html-italic">TNF-α</span> (<b>B</b>), <span class="html-italic">NF-κB</span> (<b>C</b>), and <span class="html-italic">COX-2</span> (<b>D</b>) genes in lead acetate-intoxicated rats. ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: lead acetate, <span class="html-italic">IL-6</span>: interleukin-1, <span class="html-italic">TNF-α</span>: tumor necrosis factor alpha, <span class="html-italic">NF-κB</span>: nuclear factor kappa beta, <span class="html-italic">COX-2</span>: cyclooxygenase-2. Data are expressed as mean ± S.D. (<span class="html-italic">n</span> = 10). Means that do not share a letter showed significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 9
<p>(<b>A</b>–<b>J</b>). Photomicrographs of testicular sections of the different groups. (<b>A</b>–<b>D</b>) Testis sections from the negative and ASFE control groups, which indicated normal histological structure, with normal luminal sperms (solid arrow), and spermatogonial layers (dotted arrow). (<b>E</b>–<b>H</b>) Testis sections from the PbAc-intoxicated rats with apparent morphological alteration of testicular tissue, congestion of the testicular vessels (Co), defective spermatogenic series in the seminiferous tubules (*), irregular contour of the seminiferous tubule (solid arrow), degeneration of spermatogenic epithelial series, and necrotic changes of the spermatogonial cells. (<b>I</b>,<b>J</b>) Testicular sections of PbAc-administered rats that were treated with ASFE, which had an ameliorative effect on testicular histology, restoring the architecture of the seminiferous tubule and spermatogonia cells’ integrity.</p>
Full article ">Figure 10
<p>Schematic illustration of experimental design. ASFE: <span class="html-italic">Annona squamosa</span> fruit extract, PbAc: lead acetate.</p>
Full article ">
13 pages, 4372 KiB  
Article
Tert-Butylhydroquinone Mitigates T-2-Toxin-Induced Testicular Dysfunction by Targeting Oxidative Stress, Inflammation, and Apoptosis in Rats
by Yun Chen, Xinke Zhang, Shanshan Lan, Shuping Liang, Manyu Zhang, Shuang Zhang, Yijian Liu, Li Li, Hengxi Wei and Shouquan Zhang
Toxics 2024, 12(5), 335; https://doi.org/10.3390/toxics12050335 - 5 May 2024
Viewed by 1265
Abstract
Tert-butylhydroquinone (tBHQ) has emerged as a promising candidate for mitigating the adverse effects of T-2-induced reproductive toxicity. The protective effects of tBHQ on rat sperm quality, testicular injury, apoptosis, and inflammation induced by T-2 toxin exposure were investigated. Histopathological examination of testicular tissues [...] Read more.
Tert-butylhydroquinone (tBHQ) has emerged as a promising candidate for mitigating the adverse effects of T-2-induced reproductive toxicity. The protective effects of tBHQ on rat sperm quality, testicular injury, apoptosis, and inflammation induced by T-2 toxin exposure were investigated. Histopathological examination of testicular tissues revealed severe damage in the T-2-treated group, characterized by disorganized germ cell arrangement, thinning of the convoluted seminiferous tubule walls, and significant cellular necrosis. However, tBHQ administration, either as a preventive or therapeutic measure, mitigated this structural damage. Image analysis confirmed an increase in the cross-sectional area and height of the convoluted seminiferous tubules in the tBHQ-treated groups compared to the T-2-treated group (p < 0.05), indicating tBHQ’s efficacy in alleviating testicular damage. Additionally, tBHQ treatment significantly inhibited T-2-induced apoptosis of testicular tissue cells, as evidenced by the results showing reduced apoptotic cell counts and downregulation of the BAX/BCL2 ratio and caspase-3 expression (p < 0.05). tBHQ significantly increased the concentrations of the antioxidant factors SOD, CAT, TAC, and GSH-PX. Furthermore, tBHQ attenuated the inflammatory response induced by T-2 exposure, as indicated by the decreased mRNA expression of the proinflammatory cytokines Tnf, Il1, and Il10 in testicular tissue (p < 0.05). Additionally, tBHQ treatment alleviated the decline in serum testosterone induced by the T-2 and promoted testosterone synthesis gene expression, including for the genes 17β-HSD and Cyp11a1, in rat testes (p < 0.05). These findings underscore tBHQ’s role as a therapeutic agent combatting T-2-induced reproductive toxicity, highlighting its antioxidative, anti-apoptotic, and anti-inflammatory properties. Further elucidation of tBHQ’s mechanisms of action may offer novel strategies for preventing and treating reproductive disorders induced by environmental toxins. Full article
(This article belongs to the Section Reproductive and Developmental Toxicity)
Show Figures

Figure 1

Figure 1
<p>tBHQ alleviates T-2 induced testicular injury. (<b>A</b>–<b>D</b>) are at 200× magnification, with a scale bar of 100 μm, while (<b>E</b>–<b>H</b>) are at 400× magnification, with a scale bar of 50 μm. (<b>A</b>,<b>E</b>) represent the control group, (<b>B</b>,<b>F</b>) represent the T-2 group, (<b>C</b>,<b>G</b>) represent the tBHQ+T-2 group, and (<b>D</b>,<b>H</b>) represent the M group. Testicular tissue sections stained with hematoxylin and eosin (HE) were observed under a light microscope. In the control group, germ cells were orderly arranged in various layers, with thick walls of the convoluted seminiferous tubules and abundant spermatozoa in the tubular lumens at the locations indicated by the red-boxed triangles in (<b>A</b>,<b>E</b>). Conversely, in the experimental groups, germ cells were loosely arranged and disorganized, with thinning walls of the convoluted seminiferous tubules and reduced numbers of mature spermatozoa in the tubular lumens at the locations indicated by the red-boxed triangles in (<b>B</b>–<b>D</b>,<b>F</b>,<b>H</b>). Under high magnification, observations of the T2 group revealed basal membrane disruptions in the walls of the convoluted seminiferous tubules (indicated by the black arrow in (<b>B</b>)), with numerous germ cells being detached from the tubular lumens (indicated by the black arrow in (<b>F</b>)), and the black-lined area in (<b>H</b>) defines the cross-sectional area of the convoluted seminiferous tubules. The area within the blue lines defines the luminal area. The average length of the four green double-headed arrows represents the height of the germinal epithelium. (<b>I</b>) Cross-sectional area of the convoluted seminiferous tubules. (<b>J</b>) Height of the convoluted seminiferous tubules. n = 8. * represents a significance difference, with <span class="html-italic">p</span> &lt; 0.05. ** represents a significance difference (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2
<p>tBHQ inhibits T-2-induced apoptosis of testicular cells. (<b>A</b>) TUNEL staining; (<b>B</b>) semi-quantitative analysis of apoptotic cell ratio using TUNEL staining fluorescence; (<b>C</b>) mRNA expression levels of apoptotic factors Bax, Bcl2, and Caspase-3; (<b>D</b>–<b>F</b>) protein expression levels of Bax and Bcl2, represented by grayscale values; (<b>E</b>–<b>G</b>) protein expression levels of Caspase-3, represented by grayscale values. n = 10. * represents a significant difference, with <span class="html-italic">p</span> &lt; 0.05. ** represents a significant difference, with <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>tBHQ alleviates the inflammatory response induced by T-2. (<b>A</b>) mRNA expression levels of inflammatory factors; (<b>B</b>,<b>D</b>) protein expression levels and phosphorylation of JAK, represented by grayscale values; (<b>C</b>,<b>E</b>) protein expression levels and phosphorylation of STAT3, represented by grayscale values. n = 8. * represents a significant difference, with <span class="html-italic">p</span> &lt; 0.05. ** represents a significant difference, with <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>tBHQ Attenuates T-2-induced decrease in serum testosterone levels and promotes expression of testosterone synthesis genes. (<b>A</b>) Concentrations of Gnrh, FSH, and LH in rat serum. (<b>B</b>) Concentration of testosterone in serum. (<b>C</b>) mRNA expression levels of steroid hormone synthesis genes. n = 10. * represents a significant difference, with <span class="html-italic">p</span> &lt; 0.05. ** represents a significant difference for <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
Back to TopTop