1
|
Yadav S, Yadav A, Mishra RK. Chronic unpredictable stress exposure disrupts testicular function by modulating germ cell-junctional dynamics and Nrf2/HO-1/IKKβ/NF-κB pathway. Reprod Toxicol 2025; 132:108845. [PMID: 39884400 DOI: 10.1016/j.reprotox.2025.108845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/31/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The unpredictable nature of stress complicates understanding its relationship with male infertility. In this study, we investigated testicular germ cell and junctional dynamics in male mice following exposure to chronic unpredictable stress (CUS). Adult Parkes male mice were exposed to CUS for 35 days (one complete spermatogenic cycle), with a random stressor (restraint stress, water deprivation, food deprivation, light flashing, wet bedding, cage shaking, or cage tilting) applied once per day in an intermittent and unpredictable manner to avoid repeating the same stimulus on consecutive days. CUS exposure caused behavioral alterations in mice, as observed through the forced swim test and the tail suspension test. CUS inhibited testosterone biosynthesis by decreasing steroidogenic markers (SF-1, StAR, 3β-HSD, and 17β-HSD). It also resulted in altered oxido-inflammatory and apoptotic markers, including increased LPO, Caspase-3, IKKβ, and NF-κB, along with decreased Nrf2, HO-1, SOD, and catalase in the testis. CUS exposure reduced 1 C and 4 C germ cell populations and decreased germ cell ratios (1 C:2 C, 4 C:2 C, and 4 C:S-phase), impairing sperm development. CUS disrupted meiosis initiation, chromosomal synapsis, and germ cell maintenance by reducing Stra8, SYCP3, and Piwil1 expression in the testis. It also adversely affected blood-testis barrier markers, such as ZO-1 and connexin43. These changes led to altered testicular histomorphology, reduced daily sperm production, and disrupted germ cell dynamics. The findings suggest that CUS inhibits steroidogenesis and perturbs the Nrf2/HO-1/IKKβ/NF-κB oxido-inflammatory pathway. This leads to disrupted germ cell dynamics, compromised blood-testis barrier integrity, altered histomorphology, and reduced sperm production, collectively resulting in testicular dysfunction.
Collapse
Affiliation(s)
- Shubhanshu Yadav
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Anupam Yadav
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Raghav Kumar Mishra
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| |
Collapse
|
2
|
Pavani R, Venkaiah K, Prakasam PG, Dirisala VR, Krishna PG, Kishori B, Sainath SB. Protective Effects of Resveratrol Against Perfluorooctanoic Acid-Induced Testicular and Epididymal Toxicity in Adult Rats Exposed During Their Prepubertal Period. TOXICS 2025; 13:111. [PMID: 39997926 PMCID: PMC11860439 DOI: 10.3390/toxics13020111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 02/26/2025]
Abstract
The antioxidant properties of resveratrol (RES) against oxidative toxicity induced by testicular toxicants are well documented. The current study aimed to investigate the probable beneficial role of RES on male reproduction in adult rats following prepubertal exposure to perfluorooctanoic acid (PFOA). Healthy rats of the Wistar strain (23 days old) were allocated into four groups. Rats in group I did not receive any treatment, while rats in groups II, III, and IV received RES, PFOA, and RES + PFOA, respectively, between days 23 and 56 and were monitored for up to 90 days. Exposure to PFOA resulted in a significant reduction in spermiogram parameters, testicular 3β- and 17β-HSD activity levels, and circulatory levels of testosterone. A significant elevation in LPx, PCs, H2O2, and O2-, associated with a concomitant reduction in SOD, CAT, GPx, GR, and GSH, was noticed in the testes, as well as region-specific changes in pro- and antioxidants in the epididymides of exposed rats compared to controls. A significant increase in serum FSH and LH, testicular cholesterol levels, and caspase-3 activity was observed in PFOA-exposed rats compared to controls. Histological analysis revealed that the integrity of the testes was deteriorated in PFOA-exposed rats. Transcriptomic profiling of the testes and epididymides revealed 98 and 611 altered genes, respectively. In the testes, apoptosis and glutathione pathways were disrupted, while in the epididymides, glutathione and bile secretion pathways were altered in PFOA-exposed rats. PFOA exposure resulted in the down-regulation in the testes of 17β-HSD, StAR, nfe2l2, ar, Lhcgr, and mRNA levels, associated with the up-regulation of casp3 mRNA, and down-regulation of alpha 1 adrenoceptor, muscarinic choline receptor 3, and androgen receptor in the epididymides of exposed rats compared to the controls. These events might lead to male infertility in PFOA-exposed rats. In contrast, restoration of selected reproductive variables was observed in RES plus PFOA-exposed rats compared to rats exposed to PFOA alone. Taken together, we postulate that prepubertal exposure to PFOA triggered oxidative damage and altered genes in the testes and epididymides, leading to suppressed male reproductive health in adult rats, while RES, with its steroidogenic, antiapoptotic, and antioxidant effects, restored PFOA-induced fertility potential in rats.
Collapse
Affiliation(s)
- R. Pavani
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, India; (R.P.); (K.V.); (P.G.P.)
| | - K. Venkaiah
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, India; (R.P.); (K.V.); (P.G.P.)
| | - P. Gnana Prakasam
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, India; (R.P.); (K.V.); (P.G.P.)
| | - Vijaya R. Dirisala
- Department of Biotechnology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Guntur 522213, India;
| | - P. Gopi Krishna
- Department of Zoology, Vikrama Simhapuri University PG Centre, Kavali 524201, India;
| | - B. Kishori
- Department of Biotechnology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, India;
| | - S. B. Sainath
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524324, India; (R.P.); (K.V.); (P.G.P.)
- Department of Food Technology, Vikrama Simhapuri University, Nellore 524324, India
| |
Collapse
|
3
|
Zhong J, Zhang L, Chen K, Yuan X, Cui Z, Tang S, Zheng F, Li Y, Héroux P, Wu Y, Xia D. Environmentally relevant concentration PFNA promotes degradation of SMAD7 to drive progression of ovarian cancer via TGF-β/SMADs signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116907. [PMID: 39205352 DOI: 10.1016/j.ecoenv.2024.116907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Perfluorononanoic acid (PFNA), an acknowledged environmental endocrine disruptor, is increasingly utilized as a substitute for perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA). Despite its growing use, limited research has been conducted to investigate its potential impact on tumorigenesis and progression, and the potential molecular mechanisms. Earlier studies linked perfluoroalkyl and polyfluoroalkyl substances (PFAS) exposure to breast and gynecological cancer progression in humans, lacking a clear understanding of the underlying mechanisms, notably in ovarian cancer. Our investigation into PFNA's effects at environmental concentrations (0.25-2 mM) showed no significant impact on cell proliferation but a notable increase in invasion and migration of ovarian cancer cells. This led to alterations in epithelial-mesenchymal transition (EMT) markers, including Claudin1, Vimentin, and Snail. Notably, PFNA exposure activated the TGF-β/SMADs signaling pathway. Crucially, SMAD7 degradation through the ubiquitin-proteasome system emerged as PFNA's pivotal molecular target for inducing EMT, corroborated in mouse models. In summary, this study presented evidence that environmentally relevant concentrations of PFNA could induce SMAD7 degradation via the proteasome pathway, subsequently activating the TGF-β/SMADs signaling pathway, and promoting EMT in ovarian cancer. These results illuminated the association between PFNA exposure and metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Jiamin Zhong
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lihuan Zhang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310003, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyan Cui
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Song Tang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Zheng
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China
| | - Ying Li
- Department of Epidemiology, Biostatistics and Occupational Health, 2001 McGill University, Suite 1200, Montreal H3A 1G1, Canada
| | - Paul Héroux
- Department of Epidemiology, Biostatistics and Occupational Health, 2001 McGill University, Suite 1200, Montreal H3A 1G1, Canada
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Forthun IH, Roelants M, Knutsen HK, Haug LS, Iszatt N, Schell LM, Jugessur A, Bjerknes R, Oehme NB, Madsen A, Bruserud IS, Juliusson PB. Exposure to Per- and Polyfluoroalkyl Substances and Timing of Puberty in Norwegian Boys: Data from the Bergen Growth Study 2. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:16336-16346. [PMID: 39226441 PMCID: PMC11411722 DOI: 10.1021/acs.est.4c06062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are widespread environmental contaminants with endocrine-disruptive properties. Their impact on puberty in boys is unclear. In this cross-sectional study, we investigated the association between PFAS exposure and pubertal timing in 300 Norwegian boys (9-16 years), enrolled in the Bergen Growth Study 2 during 2016. We measured 19 PFAS in serum samples and used objective pubertal markers, including ultrasound-measured testicular volume (USTV), Tanner staging of pubic hair development, and serum levels of testosterone, luteinizing hormone, and follicle-stimulating hormone. In addition to logistic regression of single pollutants and the sum of PFAS, Bayesian and elastic net regression were used to estimate the contribution of the individual PFAS. Higher levels of the sum of perfluorooctanesulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), and perfluorohexanesulfonic acid (PFHxS) were associated with later pubertal onset according to USTV (age-adjusted odds ratio (AOR): 2.20, 95% confidence interval (CI): 1.29, 3.93) and testosterone level (AOR: 2.35, 95% CI: 1.34, 4.36). Bayesian modeling showed that higher levels of PFNA and PFHxS were associated with later pubertal onset by USTV, while higher levels of PFNA and perfluoroundecanoic acid (PFUnDA) were associated with later pubertal onset by testosterone level. Our findings indicate that certain PFAS were associated with delay in male pubertal onset.
Collapse
Affiliation(s)
- Ingvild Halsør Forthun
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, 5021 Bergen, Norway
| | - Mathieu Roelants
- Department of Public Health and Primary Care, Centre for Environment and Health KU Leuven, 3000 Leuven, Belgium
| | - Helle Katrine Knutsen
- Department of Food Safety, Norwegian Institute of Public Health, 0213 Oslo, Norway
- Center for Sustainable Diets, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | - Line Småstuen Haug
- Department of Food Safety, Norwegian Institute of Public Health, 0213 Oslo, Norway
- Center for Sustainable Diets, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | - Nina Iszatt
- Department of Food Safety, Norwegian Institute of Public Health, 0213 Oslo, Norway
- Center for Sustainable Diets, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | - Lawrence M Schell
- Department of Epidemiology and Biostatistics, University at Albany, Albany, New York 12144, United States
| | - Astanand Jugessur
- Centre for Fertility and Health, Norwegian Institute of Public Health, 0213 Oslo, Norway
- Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway
| | - Robert Bjerknes
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ninnie B Oehme
- Children and Youth Clinic, Haukeland University Hospital, 5021 Bergen, Norway
| | - Andre Madsen
- Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5021 Bergen, Norway
| | | | - Petur Benedikt Juliusson
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Health Registry Research and Development, Norwegian Institute of Public Health, 5808 Bergen, Norway
| |
Collapse
|
5
|
Ku KB, Chae J, Park WH, La J, Lee SS, Lee HK. Assessment of immunopathological responses of a novel non-chemical biocide in C57BL/6 for safe disinfection usage. Lab Anim Res 2024; 40:28. [PMID: 39135094 PMCID: PMC11320990 DOI: 10.1186/s42826-024-00214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Water electrospray technology has been developed and extensively studied for its physical properties and potential application as a non-chemical biocide against airborne pathogens. However, there are still concerns regarding the safety and potential toxicity of inhaling water electrospray (WE) particles. To address these potential hazards and offer insights into the impact of WE on humans, we analyzed the immunopathological response to WE by employing an intranasal challenge C57BL/6 mouse model. This analysis aimed to compare the effects of WE with those of sodium hypochlorite (SH), a well-known biocidal agent. RESULTS The study findings suggest that the WE did not trigger any pathological immune reactions in the intranasal-challenged C57BL/6 mouse model. Mice challenged with WE did not experience body weight loss, and there was no increase in inflammatory cytokine production compared to SH-treated mice. Histopathological analysis revealed that WE did not cause any damage to the lung tissue. In contrast, mice treated with SH exhibited significant lung tissue damage, characterized by the infiltration of neutrophils and eosinophils. Transcriptomic analysis of lung tissue further confirmed the absence of a pathological immune response in mice treated with WE compared to those treated with SH. Upon intranasal challenge with WE, the C57BL/6 mouse model did not show any evidence of immunopathological damage. CONCLUSIONS The results of this study suggest that WE is a safe technology for disinfecting airborne pathogens. It demonstrated little to no effect on immune system activation and pathological outcomes in the intranasal challenge C57BL/6 mouse model. These findings not only support the potential use of WE as an effective and safe method for air disinfection but also highlight the value of the intranasal challenge of the C57BL/6 mouse model in providing significant immunopathological insights for assessing the inhalation of novel materials for potential use.
Collapse
Affiliation(s)
- Keun Bon Ku
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Center for Infectious Disease Vaccine and Diagnosis Innovation, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jihwan Chae
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Won Hyung Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jeongwoo La
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Seung S Lee
- Department of Mechanical Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
6
|
Rajpoot A, Yadav K, Yadav A, Mishra RK. Shilajit mitigates chemotherapeutic drug-induced testicular toxicity: Study on testicular germ cell dynamics, steroidogenesis modulation, and Nrf-2/Keap-1 signaling. J Ayurveda Integr Med 2024; 15:100930. [PMID: 39121783 PMCID: PMC11362644 DOI: 10.1016/j.jaim.2024.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/27/2024] [Accepted: 04/03/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Medications, including chemotherapeutic drugs, contribute to male infertility as external factors by inducing oxidative stress in testicular cells. Shilajit is a naturally occurring bioactive antioxidant used in Ayurvedic medicine to treat a variety of ailments. OBJECTIVE This study examines the potential of Shilajit to counteract the negative effects of the chemotherapeutic drug cyclophosphamide (CPA) on testicular germ cell dynamics. MATERIAL AND METHODS Male Parkes mice received single intraperitoneal CPA injection (200 mg/kg BW) on day one, followed by daily supplementation of Shilajit (100 and 200 mg/kg BW) for one spermatogenic cycle. RESULTS CPA adversely affected testicular germ cell dynamics by inhibiting the conversion of spermatogonia-to-spermatids, altering testicular histoarchitecture, impairing Sertoli cell function and testicular steroidogenesis, and disturbing the testicular oxido-apoptotic balance. Shilajit supplementation restores testicular germ cell dynamics in CPA-exposed mice, as evidenced by improved histoarchitecture of the testis. Shilajit improves testicular daily production and sperm quality by promoting the conversion of spermatogonia (2C) into spermatids (1C), stimulating germ cell proliferation (PCNA), improving Sertoli cell function (N-Cadherin and β-Catenin), and maintaining the Bax/Bcl2 ratio. Additionally, Shilajit enhances testosterone biosynthesis by activating enzymes like 3β-HSD, and 17β-HSD. Shilajit also reduces testicular oxidative stress by increasing antioxidant enzyme activity (SOD) and decreasing lipid peroxidation (LPO). These effects are mediated by upregulation of the antioxidant protein Nrf-2 and downregulation of Keap-1. CONCLUSION The findings underscore the potent androgenic and antioxidant characteristics of Shilajit, as well as its ability to enhance fertility in cases of testicular damage caused by chemotherapeutic drugs.
Collapse
Affiliation(s)
- Arti Rajpoot
- Male Reproductive Physiology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Kiran Yadav
- Male Reproductive Physiology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Anupam Yadav
- Male Reproductive Physiology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Raghav Kumar Mishra
- Male Reproductive Physiology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
7
|
Hassan YF, Shabaan DA. Effect of N-acetylcysteine on hair follicle changes in mouse model of cyclophosphamide-induced alopecia: histological and biochemical study. Histochem Cell Biol 2024; 161:477-491. [PMID: 38641701 PMCID: PMC11162382 DOI: 10.1007/s00418-024-02282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/21/2024]
Abstract
Chemotherapy-induced alopecia (CIA) represents one of the most severe side effects of chemotherapy, which forces some patients to reject cancer treatment. The exact pathophysiological mechanisms of CIA are not clearly understood, which makes it difficult to discover efficient preventive or therapeutic procedures for this adverse effect. N-acetylcysteine (NAC) has a strong antioxidant activity as it stimulates glutathione synthesis and acts as an oxygen radical scavenger. The current study tried to investigate the efficacy of NAC in preserving biochemical parameters and hair follicle structure against cyclophosphamide (CYP) administration. In total, 40 adult female C57BL/6 mice were induced to enter anagen by depilation (day 0) and divided into four groups: group I (control), group II (CYP) received a single dose of CYP [150 mg/kg body weight (B.W.)/intraperitoneal injection (IP)] at day 9, group III (CYP & NAC) received a single dose of CYP at day 9 as well as NAC (500 mg/kg B.W./day/IP) from day 6-16, and group IV (NAC) received NAC from day 6-16. CYP administration in group II induced an increase in malondialdehyde (MDA), decrease in superoxide dismutase (SOD), histological hair follicle dystrophy, disruption of follicular melanogenesis, overexpression of p53, and loss of ki67 immunoreactivity. NAC coadministration in group III reversed CYP-induced alterations in the biochemical parameters and preserved hair follicle structure, typical follicular melanin distribution as well as normal pattern of p53 and ki67 expression. These findings indicated that NAC could be used as an efficient and safe therapeutic option for hair loss induced by chemotherapy.
Collapse
Affiliation(s)
- Yomna F Hassan
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Dalia A Shabaan
- Medical Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
8
|
Zheng X, Chen J, Kang L, Wei Y, Wu Y, Hong Y, Wang X, Li D, Shen L, Long C, Wei G, Wu S. Prepubertal exposure to copper oxide nanoparticles induces Leydig cell injury with steroidogenesis disorders in mouse testes. Biochem Biophys Res Commun 2023; 654:62-72. [PMID: 36889036 DOI: 10.1016/j.bbrc.2023.02.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 02/27/2023]
Abstract
Copper oxide nanoparticles (CuONPs) are metallic multifunctional nanoparticles with good conductive, catalytic and antibacterial characteristics that have shown to cause reproductive dysfunction. However, the toxic effect and potential mechanisms of prepubertal exposure to CuONPs on male testicular development have not been clarified. In this study, healthy male C57BL/6 mice received 0, 10, and 25 mg/kg/d CuONPs by oral gavage for 2 weeks (postnatal day 22-35). The testicular weight was decreased, testicular histology was disturbed and the number of Leydig cells was reduced in all CuONPs-exposure groups. Transcriptome profiling suggested steroidogenesis was impaired after exposure to CuONPs. The steroidogenesis-related genes mRNA expression level, concentration of serum steroids hormones and the HSD17B3-, STAR- and CYP11A1-positive Leydig cell numbers were dramatically reduced. In vitro, we exposed TM3 Leydig cells to CuONPs. Bioinformatic analysis, flow cytometry analysis and western blotting analysis confirmed that CuONPs can dramatically reduce Leydig cells viability, enhance apoptosis, trigger cell cycle arrest and reduce cell testosterone levels. U0126 (ERK1/2 inhibitor) significantly reversed TM3 Leydig cells injury and testosterone level decrease induced by CuONPs. These outcomes indicate that CuONPs exposure activates the ERK1/2 signaling pathway, which further promotes apoptosis and cell cycle arrest in TM3 Leydig cells, and ultimately leads to Leydig cells injury and steroidogenesis disorders.
Collapse
Affiliation(s)
- Xiangqin Zheng
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Jiadong Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Lian Kang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Yuhao Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Xia Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Dinggang Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Lianju Shen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chunlan Long
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.
| |
Collapse
|
9
|
Wang T, Xu H, Guo Y, Li Z, Ye H, Wu L, Guo Y, Wang D. Perfluorodecanoic acid promotes adipogenesis via NLRP3 inflammasome-mediated pathway in HepG2 and 3T3-L1 cells. Food Chem Toxicol 2022; 171:113520. [PMID: 36423729 DOI: 10.1016/j.fct.2022.113520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022]
Abstract
Perfluorodecanoic acid (PFDA) is a toxic persistent pollutant that is extensively used in food applications, such as food packaging and cookware. Emerging evidence indicates that PFDA exposure were associated with higher plasma triglyceride concentration in human. In contrast, it is unknown how PFDA might affect adipogenesis. To explore the effects and underlying mechanisms of PFDA on lipid metabolism in this study, both HepG2 cells and 3T3-L1 differentiation model were used. The results showed that PFDA promoted the cellular triglyceride accumulation and triglyceride content in concentration-dependent manners. Furthermore, PFDA activated the NLRP3 inflammasome, which is crucial for the induction of lipogenic genes expression including fatty acid synthase (FAS), hydroxymethylglutaryl coenzyme A synthase (HMGCS), and stearoyl-CoA desaturase 1 (SCD1). Additionally, PFDA-induced adipogenesis was abolished by caspase-1 inhibitor and siNLRP3 in HepG2 cells. Moreover, after PFDA treatment, the expression of SREBP1, an important regulator of lipid metabolism, was increased, as well as its target genes, and PFDA-induced SREBP1 enhanced expression can be abolished by caspase-1 inhibitor and siNLRP3 as well. Together, these results provide to understanding of the potential health implications of exposure to PFDA on lipid accumulation, and suggest that PFDA can promote adipogenesis via an NLRP3 inflammasome-mediated SREBP1 pathway.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Yu Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Zhanming Li
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Hua Ye
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China
| | - Liang Wu
- School of Medicine, Jiangsu University, 212013, Zhenjiang, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100, Zhenjiang, China.
| |
Collapse
|
10
|
Gundacker C, Audouze K, Widhalm R, Granitzer S, Forsthuber M, Jornod F, Wielsøe M, Long M, Halldórsson TI, Uhl M, Bonefeld-Jørgensen EC. Reduced Birth Weight and Exposure to Per- and Polyfluoroalkyl Substances: A Review of Possible Underlying Mechanisms Using the AOP-HelpFinder. TOXICS 2022; 10:toxics10110684. [PMID: 36422892 PMCID: PMC9699222 DOI: 10.3390/toxics10110684] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 05/14/2023]
Abstract
Prenatal exposure to per- and polyfluorinated substances (PFAS) may impair fetal growth. Our knowledge of the underlying mechanisms is incomplete. We used the Adverse Outcome Pathway (AOP)-helpFinder tool to search PubMed for studies published until March 2021 that examined PFAS exposure in relation to birth weight, oxidative stress, hormones/hormone receptors, or growth signaling pathways. Of these 1880 articles, 106 experimental studies remained after abstract screening. One clear finding is that PFAS are associated with oxidative stress in in vivo animal studies and in vitro studies. It appears that PFAS-induced reactive-oxygen species (ROS) generation triggers increased peroxisome proliferator-activated receptor (PPAR)γ expression and activation of growth signaling pathways, leading to hyperdifferentiation of pre-adipocytes. Fewer proliferating pre-adipocytes result in lower adipose tissue weight and in this way may reduce birth weight. PFAS may also impair fetal growth through endocrine effects. Estrogenic effects have been noted in in vivo and in vitro studies. Overall, data suggest thyroid-damaging effects of PFAS affecting thyroid hormones, thyroid hormone gene expression, and histology that are associated in animal studies with decreased body and organ weight. The effects of PFAS on the complex relationships between oxidative stress, endocrine system function, adipogenesis, and fetal growth should be further explored.
Collapse
Affiliation(s)
- Claudia Gundacker
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +43-1-40160-56503
| | - Karine Audouze
- Unit T3S, Université Paris Cité, Inserm U1124, 75006 Paris, France
| | - Raimund Widhalm
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Sebastian Granitzer
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Forsthuber
- Institute of Medical Genetics, Medical University of Vienna, 1090 Vienna, Austria
| | - Florence Jornod
- Unit T3S, Université Paris Cité, Inserm U1124, 75006 Paris, France
| | - Maria Wielsøe
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
| | - Manhai Long
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
| | - Thórhallur Ingi Halldórsson
- Faculty of Food Science and Nutrition, University of Iceland, 102 Reykjavík, Iceland
- Department of Epidemiology Research, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Maria Uhl
- Environment Agency Austria, 1090 Vienna, Austria
| | - Eva Cecilie Bonefeld-Jørgensen
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark
- Greenland Center for Health Research, Greenland University, Nuuk 3905, Greenland
| |
Collapse
|
11
|
Yadav A, Yadav K, Rajpoot A, Lal B, Mishra RK. Sub-chronic restraint stress exposure in adult rats: An insight into possible inhibitory mechanism on testicular function in relation to germ cell dynamics. Andrologia 2022; 54:e14575. [PMID: 36056817 DOI: 10.1111/and.14575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/14/2022] [Accepted: 08/21/2022] [Indexed: 11/28/2022] Open
Abstract
Psychological stress is now widely recognized as one of the major risk factors for male fertility. Its impact on the dynamics of testicular germ cells, however, has yet to be fully investigated. Therefore, we used the rat restraint stress (RS) model as a psychological stressor to assess the impact of psychological stress on testicular germ cell dynamics. Adult male SD rats were exposed to sub-chronic RS for 1.5 and 3 h per day for 30 days. The quality of cauda epididymis spermatozoa was adversely affected by RS exposure, and the frequency of spermatozoa with tail abnormalities was higher than that of spermatozoa with head abnormalities. RS exposure adversely affected testicular daily sperm production by disturbing the meiotic and post meiotic germ cell kinetics in the testis. The histomorphology of the testis was altered by loosening and vacuolization in the seminiferous epithelium, germ cell exfoliation and the presence of giant cells. Seminiferous tubules of stage I-VI and VII-VIII were severely affected in rats exposed to RS for 3 h. By interfering with steroidogenic enzymes, RS exposure disrupts testosterone biosynthesis. The testicular oxidative balance was also disturbed by RS exposure, which disrupted the levels/activities of lipid peroxidation, Nrf-2, superoxide dismutase and catalase. There was also an increase in caspase-3 activity and a decrease in the Bax-Bcl2 ratio. In conclusion, our findings suggest that psychological stressors like RS impair testicular functions in rats by disrupting germ cell dynamics, downregulating testicular androgenesis and increasing oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Anupam Yadav
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Kiran Yadav
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Arti Rajpoot
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Bechan Lal
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Raghav Kumar Mishra
- Male Reproductive Physiology Lab., Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
12
|
Hong Y, Zhou Y, Shen L, Wei Y, Long C, Fu Y, Wu H, Wang J, Wu Y, Wu S, Wei G. Exposure to DEHP induces testis toxicity and injury through the ROS/mTOR/NLRP3 signaling pathway in immature rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 227:112889. [PMID: 34649140 DOI: 10.1016/j.ecoenv.2021.112889] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 06/13/2023]
Abstract
As the most abundantly used phthalate derivative, di-(2-ethylhexyl) phthalate (DEHP) leads to reproductive disorders, especially in males. Testicular injury can be triggered when the testis is exposed to DEHP during the immature stage. However, the potential mechanism is largely unclear. In the present study, Sprague-Dawley rats were exposed to 0, 250 and 500 mg/kg/day DEHP from postnatal day (PND) 20 to PND 30. The spermatogonia cell line GC-1 and spermatocyte cell line GC-2 were exposed to different doses of monoethylhexyl phthalate (MEHP), a metabolite of DEHP. Testicular injury was observed. Oxidative stress was evaluated both in vivo and in vitro. Our results showed that after DEHP exposure, the testicular structure was damaged and spermatogenesis was disturbed. We also found that oxidative stress was increased, as indicated by the upregulation of the important factors in the antioxidant pathway. Furthermore, the expression of autophagy-related proteins was significantly downregulated. Autophagy inhibition led to activation of the pyroptosis pathway. Nucleotide-binding and oligomerisation (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3), Caspase-1 and cytokine interleukin-1β (IL-1β) were significantly upregulated. Additionally, an imbalance in self-renewal and differentiation was observed in germ cells after DEHP exposure, causing the cessation of germ cell development. In summary, these data suggest that DEHP exposure enhances oxidative stress, downregulates autophagy, induces NLRP3 inflammasome activation and subsequently triggers pyroptosis in vivo and in vitro, which provides novel insight into DEHP-related injury in immature testes in the context of pyroptosis.
Collapse
Affiliation(s)
- Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Yu Zhou
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Lianju Shen
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Chunlan Long
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Yan Fu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Huan Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Junke Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Yuhao Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China.
| |
Collapse
|
13
|
Li X, Ru S, Tian H, Zhang S, Lin Z, Gao M, Wang J. Combined exposure to environmentally relevant copper and 2,2'-dithiobis-pyridine induces significant reproductive toxicity in male guppy (Poecilia reticulata). THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 797:149131. [PMID: 34346372 DOI: 10.1016/j.scitotenv.2021.149131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Metal pyrithiones (MePTs), the most widely used biocides in antifouling paints (AFs) coated on the hulls, are usually used in combination with Cu-containing substances. In the aquatic environment, 2,2'-dithiobis-pyridine ((PS)2), the main degradation product of MePTs, and Cu usually coexist. However, their combined impacts on aquatic organisms are unclear. This study exposed male guppy (Poecilia reticulata) to an environmentally realistic concentration of Cu (10 μg/L) alone or Cu (10 μg/L) combined with 20, 200, and 2000 ng/L (PS)2 to explore their combined reproductive toxicity. The results showed that co-exposure to Cu and (PS)2 increased Cu accumulation in the fish body in a dose-dependent manner and induced obvious spermatozoon apoptosis and necrosis, which was mediated by the peroxidation and caspase activation. Compared to Cu alone, co-exposure to Cu and 200, 2000 ng/L (PS)2 significantly decreased the testosterone level and collapsed spermatogenesis, and depressed male's sexual interest and mating behavior were observed in three co-exposure groups. Moreover, co-exposure to Cu and (PS)2 increased the disturbance on cyp19a and cyp19b transcription and suppressed the "display" reproductive behavior. Eventually, co-exposure to Cu and (PS)2 caused male reproductive failure. Therefore, the concurrence of Cu and (PS)2 induced significant reproductive toxicity in male guppies and would threaten the sustainability of fish populations. Considering the extensive usage of MePTs products in the AFs, their ecological risk warrants more evaluation.
Collapse
Affiliation(s)
- Xuefu Li
- Colleges of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Shaoguo Ru
- Colleges of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Hua Tian
- Colleges of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Suqiu Zhang
- Colleges of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Zhenxian Lin
- School of Biology and Brewing Engineering, Taishan University, 525 Dongyue Street, Tai'an 271000, Shandong Province, China
| | - Ming Gao
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, Jiangsu Province, China
| | - Jun Wang
- Colleges of Marine Life Sciences, Ocean University of China, Qingdao 266003, Shandong Province, China.
| |
Collapse
|
14
|
Wang C, Jin C, Tu W, Jin Y. Maternal exposure of mice to sodium p-perfluorous nonenoxybenzene sulfonate causes endocrine disruption in both dams and offspring. Endocr J 2021; 68:1165-1177. [PMID: 33980773 DOI: 10.1507/endocrj.ej20-0781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The toxicity of certain novel perfluoroalkyl substances (PFCs) has attracted increasing attention. However, the toxic effects of sodium p-perfluorous nonenoxybenzene sulfonate (OBS) on the endocrine system have not been elucidated. In this study, OBS was added to the drinking water during the pregnancy and lactation of the healthy female mice at dietary levels of 0.0 mg/L (CON), 0.5 mg/L (OBS-L), and 5.0 mg/L (OBS-H). OBS exposure during the pregnancy and lactation resulted in the presence of OBS residues in the placenta and fetus. We also analyzed physiological and biochemical parameters and gene expression levels in mice of the F0 and F1 generations after maternal OBS exposure. The total serum cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels were significantly increased in female mice of the F0 generation. The androgen levels in the serum and the ovarian mRNA levels of androgen receptor (AR) also tended to increase after maternal OBS exposure in the F0 generation mice. Moreover, maternal OBS exposure altered the mRNA expression of endocrine-related genes in male mice of F1 generation. Notably, the serum TC and LDL-C levels were significantly increased in 8-weeks-old male mice of the F1 generation, and the serum high-density lipoprotein cholesterol (HDL-C) levels were decreased in 24-week-old male mice of the F1 generation. These results indicated that maternal OBS exposure can interfere with endocrine homeostasis in the F0 and F1 generations. Therefore, exposure to OBS during pregnancy and lactation has the potential toxic effects on the dams and male offspring, which cannot be overlooked.
Collapse
Affiliation(s)
- Caiyun Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Cuiyuan Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China
| | - Yuanxiang Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
15
|
Jiao X, Liu N, Xu Y, Qiao H. Perfluorononanoic acid impedes mouse oocyte maturation by inducing mitochondrial dysfunction and oxidative stress. Reprod Toxicol 2021; 104:58-67. [PMID: 34246765 DOI: 10.1016/j.reprotox.2021.07.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/18/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022]
Abstract
Perfluorononanoic acid (PFNA), a member of PFAS, is frequently detected in human blood and tissues, even in follicular fluid of women. The exposure of PFNA, but not PFOA and PFOS, is positively correlated with miscarriage and increased time to pregnancy. Toxicological studies indicated that PFNA exposure is associated with immunotoxicity, hepatotoxicity, developmental toxicity, and reproductive toxicity in animals. However, there is little information regarding the toxic effects of PFNA on oocyte maturation. In this study, we investigated the toxic effects of PFNA exposure on mouse oocyte maturation in vitro. Our results showed that 600 μM PFNA significantly inhibited germinal vesicle breakdown (GVBD) and polar body extrusion (PBE) in mouse oocytes. Our further study revealed that PFNA induced abnormal metaphase I (MI) spindle assembly, evidenced by malformed spindles and mislocalization of p-ERK1/2 in PFNA-treated oocytes. We also found that PFNA induced abnormal mitochondrial distribution and increased mitochondrial membrane potential. Consequently, PFNA increased reactive oxygen species (ROS) levels, leading to oxidative stress, DNA damage, and eventually early-stage apoptosis in oocytes. In addition, after 14 h culture, PFNA disrupted the formation of metaphase II (MII) spindle in most PFNA-treated oocytes with polar bodies. Collectively, our results indicate that PFNA interferes with oocyte maturation in vitro via disrupting spindle assembly, damaging mitochondrial functions, and inducing oxidative stress, DNA damage, and early-stage apoptosis.
Collapse
Affiliation(s)
- Xiaofei Jiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ning Liu
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yiding Xu
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Huanyu Qiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
16
|
Lombó M, Herráez P. The effects of endocrine disruptors on the male germline: an intergenerational health risk. Biol Rev Camb Philos Soc 2021; 96:1243-1262. [PMID: 33660399 DOI: 10.1111/brv.12701] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/22/2022]
Abstract
Environmental pollution is becoming one of the major concerns of society. Among the emerging contaminants, endocrine-disrupting chemicals (EDCs), a large group of toxicants, have been the subject of many scientific studies. Besides the capacity of these compounds to interfere with the endocrine system, they have also been reported to exert both genotoxic and epigenotoxic effects. Given that spermatogenesis is a coordinated process that requires the involvement of several steroid hormones and that entails deep changes in the chromatin, such as DNA compaction and epigenetic remodelling, it could be affected by male exposure to EDCs. A great deal of evidence highlights that these compounds have detrimental effects on male reproductive health, including alterations to sperm motility, sexual function, and gonad development. This review focuses on the consequences of paternal exposure to such chemicals for future generations, which still remain poorly known. Historically, spermatozoa have long been considered as mere vectors delivering the paternal haploid genome to the oocyte. Only recently have they been understood to harbour genetic and epigenetic information that plays a remarkable role during offspring early development and long-term health. This review examines the different modes of action by which the spermatozoa represent a key target for EDCs, and analyses the consequences of environmentally induced changes in sperm genetic and epigenetic information for subsequent generations.
Collapse
Affiliation(s)
- Marta Lombó
- Department of Animal Reproduction, INIA, Puerta de Hierro 18, Madrid, 28040, Spain
| | - Paz Herráez
- Department of Molecular Biology, Faculty of Biology, Universidad de León, Campus de Vegazana s/n, León, 24071, Spain
| |
Collapse
|
17
|
Low dose of fire retardant, 2,2',4,4'-tetrabromodiphenyl ether (BDE47), stimulates the proliferation and differentiation of progenitor Leydig cells of male rats during prepuberty. Toxicol Lett 2021; 342:6-19. [PMID: 33581290 DOI: 10.1016/j.toxlet.2021.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 01/17/2023]
Abstract
2,2',4,4'-Tetrabromodiphenyl ether (BDE47), a flame retardant, is extensively distributed in the food chain. However, whether BDE47 affects Leydig cell development during prepuberty remains unclear. BDE47 was daily gavaged to 21-day-old Sprague-Dawley male rats with 0 (corn oil), 0.1, 0.2, and 0.4 mg/kg for 14 days. BDE47 did not affect the body weight or testis weight of rats. It significantly increased serum testosterone level at 0.4 mg/kg, but decreased luteinizing hormone (LH) level without affecting estradiol level. BDE47 induced Leydig cell hyperplasia (the number of CYP11A1-positive Leydig cells increased), and up-regulated the expression of Scarb1, Star, Hsd11b1, Pcna, and Ccnd1 in the testis. BDE47 significantly reduced p53 and p21 levels but increased CCND1 level. It also markedly increased the phosphorylation of AKT1, AKT2, ERK1/2, and CREB. BDE47 significantly up-regulated the expression of Scarb1, Star, and Hsd11b1 and stimulated androgen production by immature Leydig cells from rats under basal, LH, and 8Br-cAMP stimulated conditions at 100 nM in vitro. In conclusion, BDE47 increased Leydig cell number and up-regulated the expression of Scarb1 and Star, thereby leading to increased testosterone synthesis.
Collapse
|
18
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Ceccatelli S, Cravedi J, Halldorsson TI, Haug LS, Johansson N, Knutsen HK, Rose M, Roudot A, Van Loveren H, Vollmer G, Mackay K, Riolo F, Schwerdtle T. Risk to human health related to the presence of perfluoroalkyl substances in food. EFSA J 2020; 18:e06223. [PMID: 32994824 PMCID: PMC7507523 DOI: 10.2903/j.efsa.2020.6223] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluoroalkyl substances (PFASs) in food. Based on several similar effects in animals, toxicokinetics and observed concentrations in human blood, the CONTAM Panel decided to perform the assessment for the sum of four PFASs: PFOA, PFNA, PFHxS and PFOS. These made up half of the lower bound (LB) exposure to those PFASs with available occurrence data, the remaining contribution being primarily from PFASs with short half-lives. Equal potencies were assumed for the four PFASs included in the assessment. The mean LB exposure in adolescents and adult age groups ranged from 3 to 22, the 95th percentile from 9 to 70 ng/kg body weight (bw) per week. Toddlers and 'other children' showed a twofold higher exposure. Upper bound exposure was 4- to 49-fold higher than LB levels, but the latter were considered more reliable. 'Fish meat', 'Fruit and fruit products' and 'Eggs and egg products' contributed most to the exposure. Based on available studies in animals and humans, effects on the immune system were considered the most critical for the risk assessment. From a human study, a lowest BMDL 10 of 17.5 ng/mL for the sum of the four PFASs in serum was identified for 1-year-old children. Using PBPK modelling, this serum level of 17.5 ng/mL in children was estimated to correspond to long-term maternal exposure of 0.63 ng/kg bw per day. Since accumulation over time is important, a tolerable weekly intake (TWI) of 4.4 ng/kg bw per week was established. This TWI also protects against other potential adverse effects observed in humans. Based on the estimated LB exposure, but also reported serum levels, the CONTAM Panel concluded that parts of the European population exceed this TWI, which is of concern.
Collapse
|
19
|
Zhu Q, Li H, Wen Z, Wang Y, Li X, Huang T, Mo J, Wu Y, Zhong Y, Ge RS. Perfluoroalkyl substances cause Leydig cell dysfunction as endocrine disruptors. CHEMOSPHERE 2020; 253:126764. [PMID: 32464778 DOI: 10.1016/j.chemosphere.2020.126764] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 06/11/2023]
Abstract
Perfluoroalkyl substances (PFASs) are a group of man-made organic substances. Some of PFASs have been classified as persistent organic pollutants and endocrine disruptors. They might interfere with the male sex endocrine system, causing the abnormal development of the male reproductive tract and failure of pubertal onset and infertility. The present review discusses the development and function of two generations of Leydig cells in rodents and the effects of PFASs on Leydig cell development after their exposure in gestational and postnatal periods. We also discuss human epidemiological data for the effects of PFASs on male sex hormone levels. The structure-activity relationship of PFASs on Leydig cell steroidogenesis and enzyme activities are also discussed.
Collapse
Affiliation(s)
- Qiqi Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zina Wen
- Chengdu Xi'nan Gynecological Hospital, Chengdu, Sichuan, China
| | - Yiyang Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoheng Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tongliang Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaying Mo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Zhong
- Chengdu Xi'nan Gynecological Hospital, Chengdu, Sichuan, China.
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
20
|
Kumar J, Haldar C, Verma R. Fluoride Compromises Testicular Redox Sensor, Gap Junction Protein, and Metabolic Status: Amelioration by Melatonin. Biol Trace Elem Res 2020; 196:552-564. [PMID: 31828722 DOI: 10.1007/s12011-019-01946-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
Abstract
The excess fluoride intake has been shown to adversely affect male reproductive health. The aim of the present study was to investigate the key mechanism underlying fluoride-induced testicular dysfunction and the role of melatonin as a modulator of testicular metabolic, oxidative, and inflammatory load. The present results indicated that sodium fluoride (NaF) exposure to adult male golden hamsters severely impairs reproductive physiology as evident from markedly reduced sperm count/viability, testosterone level, androgen receptor (AR), testicular glucose transporter (GLUT-1), gap junction (connexin-43), and survival (Bcl-2) protein expression. NaF exposure markedly increased testicular oxidative load, inflammatory (NF-kB/COX-2), and apoptotic (caspase-3) protein expression. However, melatonin treatment remarkably restored testicular function as evident by normal histoarchitecture, increased sperm count/viability, enhanced antioxidant enzyme activities (SOD and Catalase), and decreased lipid peroxidation (LPO) level. In addition, melatonin treatment upregulated testicular Nrf-2/HO-I, SIRT-1/ FOXO-1, and downregulated NF-kB/COX-2 expression. Further, melatonin ameliorated NaF-induced testicular metabolic stress by modulating testicular GLUT-1expression, glucose level, and LDH activity. Furthermore, melatonin treatment enhanced testicular PCNA, Bcl-2, connexin-43, and reduced caspase-3 expression. In conclusion, we propose the molecular mechanism of fluoride-induced testicular damages and ameliorative action(s) of melatonin.
Collapse
Affiliation(s)
- Jitendra Kumar
- Department of Zoology, Pineal Research Laboratory, Reproduction Biology Unit, Institute of Science, Banaras Hindu University, Varanasi, U.P., 221005, India
| | - Chandana Haldar
- Department of Zoology, Pineal Research Laboratory, Reproduction Biology Unit, Institute of Science, Banaras Hindu University, Varanasi, U.P., 221005, India
| | - Rakesh Verma
- Department of Zoology, Pineal Research Laboratory, Reproduction Biology Unit, Institute of Science, Banaras Hindu University, Varanasi, U.P., 221005, India.
| |
Collapse
|
21
|
Selvaraju V, Baskaran S, Agarwal A, Henkel R. Environmental contaminants and male infertility: Effects and mechanisms. Andrologia 2020; 53:e13646. [PMID: 32447772 DOI: 10.1111/and.13646] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
The escalating prevalence of male infertility and decreasing trend in sperm quality have been correlated with rapid industrialisation and the associated discharge of an excess of synthetic substances into the environment. Humans are inevitably exposed to these ubiquitously distributed environmental contaminants, which possess the ability to intervene with the growth and function of male reproductive organs. Several epidemiological reports have correlated the blood and seminal levels of environmental contaminants with poor sperm quality. Numerous in vivo and in vitro studies have been conducted to investigate the effect of various environmental contaminants on spermatogenesis, steroidogenesis, Sertoli cells, blood-testis barrier, epididymis and sperm functions. The reported reprotoxic effects include alterations in the spermatogenic cycle, increased germ cell apoptosis, inhibition of steroidogenesis, decreased Leydig cell viability, impairment of Sertoli cell structure and function, altered expression of steroid receptors, increased permeability of blood-testis barrier, induction of peroxidative and epigenetic alterations in spermatozoa resulting in poor sperm quality and function. In light of recent scientific reports, this review discusses the effects of environmental contaminants on the male reproductive function and the possible mechanisms of action.
Collapse
Affiliation(s)
- Vaithinathan Selvaraju
- Department of Nutrition, Dietetics and Hospitality Management, Auburn University, Auburn, AL, USA
| | - Saradha Baskaran
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ralf Henkel
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.,Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
22
|
Zhao TX, Wang JK, Shen LJ, Long CL, Liu B, Wei Y, Han LD, Wei YX, Wu SD, Wei GH. Increased m6A RNA modification is related to the inhibition of the Nrf2-mediated antioxidant response in di-(2-ethylhexyl) phthalate-induced prepubertal testicular injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 259:113911. [PMID: 31923814 DOI: 10.1016/j.envpol.2020.113911] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/03/2019] [Accepted: 01/01/2020] [Indexed: 06/10/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a common environmental endocrine disrupting chemical that may induce male reproductive disorders. Exposure to DEHP at a prepubertal stage could lead to prepubertal testicular injury, but the underlying mechanisms remain unclear. In this study, we exposed Sprague-Dawley rats to 0, 250, and 500 mg DEHP per kg body weight per day at the prepuberty stage from postnatal day 22 (PND 22) to PND 35 by oral gavage. Testicular injury and oxidative stress were evaluated, and the levels of 6-methyladenosine (m6A) modification and expression of modulator genes for RNA methylation were measured in testes. Furthermore, m6A modification of the important antioxidant transcription factor Nrf2 was analyzed using methylated RNA immunoprecipitation qPCR. Our results show that DEHP worsened testicular histology, decreased testosterone concentrations, downregulated expression of spermatogenesis inducers, enhanced oxidative stress, inhibited the Nrf2-mediated antioxidant pathway, and increased apoptosis in testes. Additionally, DEHP increased global levels of m6A RNA modification and altered the expression of two important RNA methylation modulator genes, FTO and YTHDC2. Moreover, m6A modification of Nrf2 mRNA increased upon DEHP exposure. Overall, these findings link oxidative stress imbalance with epigenetic effects of DEHP toxicity and provide insight into the testicular toxicity of DEHP from the new perspective of m6A modification.
Collapse
Affiliation(s)
- Tian-Xin Zhao
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Jun-Ke Wang
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Lian-Ju Shen
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Chun-Lan Long
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Bin Liu
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Yi Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Lin-Dong Han
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Yue-Xin Wei
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| | - Sheng-De Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China.
| | - Guang-Hui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, PR China
| |
Collapse
|
23
|
Singh S, Singh SK. Acute exposure to perfluorononanoic acid in prepubertal mice: Effect on germ cell dynamics and an insight into the possible mechanisms of its inhibitory action on testicular functions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 183:109499. [PMID: 31398581 DOI: 10.1016/j.ecoenv.2019.109499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 05/15/2023]
Abstract
Perfluoroalkyl acids (PFAAs) are anthropogenic compounds used globally in a variety of commercial products. Perfluorononanoic acid (PFNA), a member of PFAAs, is detected in human blood and this has been reported to cause hepatotoxic, immunotoxic, and developmental and testicular toxic effects in laboratory animals. We have recently shown that the acute exposure to PFNA in prepubertal Parkes (P) mice impairs spermatogenesis by inducing oxidative stress and inhibiting testosterone biosynthesis in the testis. The present study was aimed to examine the effect of acute exposure to PFNA in prepubertal P mice on germ cell dynamics and to understand the possible mechanisms of action of this compound on testicular functions. PFNA (2 and 5 mg/kg body weight) was orally administered to male mice for 14 days from postnatal day 25-38. The treatment caused a decrease in overall germ cell transformation. The results also reveal that impairment in testicular functions in treated mice is associated with alterations in cholesterol and glucose homeostasis; further, an inhibition in expressions of growth hormone receptor (GHR), insulin-like growth factor-1 (IGF-1), insulin-like growth factor-1 receptor (IGF-1R), androgen receptor (AR), phosphorylated mammalian target of rapamycin (p-mTOR) and peroxisome proliferator activated receptor α (PPAR α) in the testis is also implicated in this action. The findings thus suggest involvement of multiple factors which altogether contribute to the alterations in spermatogenic process and testosterone production following acute exposure to PFNA in prepubertal mice.
Collapse
Affiliation(s)
- Shilpi Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shio Kumar Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
24
|
Luo Q, Li Y, Huang C, Cheng D, Ma W, Xia Y, Liu W, Chen Z. Soy Isoflavones Improve the Spermatogenic Defects in Diet-Induced Obesity Rats through Nrf2/HO-1 Pathway. Molecules 2019; 24:E2966. [PMID: 31443330 PMCID: PMC6719105 DOI: 10.3390/molecules24162966] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 01/12/2023] Open
Abstract
Soy isoflavones (SIF) are biologically active compounds of non-steroidal and phenolic properties that are richly present in soybeans, which can reduce the body weight and blood lipids of obese animals. Recently, SIF have been reported to affect reproductive ability in obese male rats. However, the specific mechanism has not been well defined. The aim of the current study was to study the possible mechanisms for the effect of SIF administration on obesity induced spermatogenic defects. Obese rats model induced by high-fat diets were established and gavage treated with 0, 50,150 or 450 mg of SIF/kg body weight/day for 4 weeks. Here, our research shows that obesity resulted in spermatogenic degeneration, imbalance of reproductive hormone, testicular oxidative stress and germ cell apoptosis, whereas evidently recovery effects were observed at 150 and 450 mg/kg SIF. We also have discovered that 150 and 450 mg/kg SIF can activate Nrf2/HO-1 pathway in control of Bcl-2, BAX and cleaved caspase-3 expression with implications in antioxidant protection. Our study indicates the potential mechanism of SIF regulating spermatogenic function in obese rats, and provides a scientific experimental basis for the regulation of biological function of obese male reproductive system by SIF.
Collapse
Affiliation(s)
- Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yifan Li
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dongjing Cheng
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenjing Ma
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
25
|
Singh S, Singh SK. Effect of gestational exposure to perfluorononanoic acid on neonatal mice testes. J Appl Toxicol 2019; 39:1663-1671. [PMID: 31389053 DOI: 10.1002/jat.3883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/24/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Perfluoroalkyl acids (PFAAs) are widely used in commercial products and are found in many goods of daily use. Perfluorononanoic acid (PFNA) is one of the PFAAs that possesses endocrine disrupting properties and we have recently shown that PFNA affects testicular functions in Parkes mice. Exposure to environmental endocrine disruptors during fetal life is believed to affect gonadal development and they might produce reproductive abnormalities in males. Therefore, the present study examined the effect of gestational exposure to PFNA on the testes of neonatal mice offspring. Pregnant Parkes mice were orally administered PFNA (2 and 5 mg/kg body weight) or distilled water from gestational day 12 until parturition. Male pups were killed on postnatal day 3. PFNA treatment decreased testosterone biosynthesis by inhibiting expression of steroidogenic acute regulatory protein, cytochrome P450scc, and 3β- and 17β-hydroxysteroid dehydrogenase; proliferation of testicular cells was also affected in treated mice. Furthermore, a marked decrease in expression of Wilms tumor 1, steroidogenic factor 1 and insulin-like factor 3 was noted in neonatal mice testes, indicating that the PFNA treatment may affect the development of the testis. Moreover, observation of the dose-related expression of anti-müllerian hormone and c-Kit in neonatal mice testes is also suggestive of an interference with gonadal development by PFNA exposure. In conclusion, the results suggest that the gestational exposure to PFNA decreased testosterone biosynthesis and altered the expression of critical factors involved in the development of the testis, thereby advocating a potential risk of PFNA to male reproductive health.
Collapse
Affiliation(s)
- Shilpi Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shio Kumar Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
26
|
Fort DJ, Mathis MB, Fort CE, Fort HM, Fort TD, Guiney PD, Weeks JA. Effect of perfluorooctanesulfonate exposure on steroid hormone levels and steroidogenic enzyme activities in juvenile Silurana tropicalis. J Appl Toxicol 2019; 39:1066-1078. [PMID: 30847954 DOI: 10.1002/jat.3794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 01/02/2023]
Abstract
The impact of the perfluoro-chemical, perfluorooctanesulfonate (PFOS), on gonadal steroidogenesis during sexual differentiation in Silurana tropicalis was examined because of its ubiquity in the environment, bioaccumulative nature and potential to disturb endocrine activity. A partial life cycle study exposing S. tropicalis to varying concentrations of PFOS 0.06, 0.13, 0.25, 0.50 and 1.0 mg PFOS/L [nominal]) was conducted. Gonad and plasma samples were collected from juvenile control specimens and organisms exposed to PFOS from early embryo through 150 days post-metamorphosis. Gonad CYP17, aromatase and 5α-reductase activities were measured. Plasma estradiol, testosterone, dihydrotestosterone (DHT) and gonadal testosterone were measured in both males and females. Increased plasma DHT and gonadal testosterone were found in PFOS-treated juvenile male S. tropicalis compared to controls. Decreased plasma estradiol, but not testosterone, was detected in PFOS-treated female S. tropicalis compared to controls. Plasma DHT was not detected and an increase in gonadal testosterone was detected in PFOS-treated female frogs. Female S. tropicalis exposed to PFOS exhibited a concentration-related decrease in the mean aromatase activity, but not 5α-reductase. PFOS exposure in male frogs induced a concentration-related increase in 5α-reductase activity, but did not alter aromatase activity compared to control frogs. A concentration-related increase in CYP 17,20-lyase activity, but not 17-hydroxylase activity, was found in both female and male S. tropicalis exposed to PFOS.
Collapse
Affiliation(s)
| | | | | | - Hayley M Fort
- Fort Environmental Laboratories, Stillwater, OK, USA
| | - Troy D Fort
- Fort Environmental Laboratories, Stillwater, OK, USA
| | | | | |
Collapse
|