1
|
Fan Z, Hong R, Li S, Kong L, Zhou Q, Ma T, Chen H, Pan C. Embryonic exposure to GenX causes reproductive toxicity by disrupting the formation of the blood-testis barrier in mouse offspring. Toxicology 2025; 515:154161. [PMID: 40268268 DOI: 10.1016/j.tox.2025.154161] [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: 02/12/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
As a replacement for perfluorooctanoic acid, hexafluoropropylene oxide dimer acid, commercially referred to as "GenX", has attracted significant attention. However, a comprehensive understanding of the reproductive systems of male offspring exposed to GenX is lacking. This study aimed to investigate how embryonic exposure to GenX affects the reproductive development of male offspring and the underlying mechanisms. We administered GenX daily via gavage (2 mg/kg body weight/day) to the mice from day 12.5 of pregnancy until delivery. Our results suggested that embryonic exposure to GenX led to delayed onset of puberty in male offspring, with destruction of the testicular structure, disruption of the blood-testis barrier, decreased serum testosterone levels, decreased sperm count, impaired sperm motility, and increased rates of sperm abnormalities. We investigated the mechanism of blood-testis barrier breakdown in vitro by treating Sertoli cells (TM4) with GenX. GenX exposure caused the accumulation of senescent TM4 cells, decreased their glutathione (GSH) levels, and increased their oxidized glutathione levels. GenX inhibited glutaminase activity in TM4 cells, leading to decreased GSH synthesis, increased intracellular oxidative stress, and subsequent TM4 cell senescence, ultimately compromising the blood-testis barrier. Our findings indicated that embryonic exposure to GenX may cause Sertoli cell senescence by altering glutamine metabolism, disrupting the blood-testis barrier, and resulting in abnormal reproductive development in male offspring.
Collapse
Affiliation(s)
- Zhencheng Fan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Runyang Hong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Key Lab of Non-Coding RNA Basic and Clinical Translational Research, Yangzhou University, Yangzhou, China
| | - Shuhao Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Key Lab of Non-Coding RNA Basic and Clinical Translational Research, Yangzhou University, Yangzhou, China
| | - Liang Kong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Key Lab of Non-Coding RNA Basic and Clinical Translational Research, Yangzhou University, Yangzhou, China
| | - Qiyue Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Key Lab of Non-Coding RNA Basic and Clinical Translational Research, Yangzhou University, Yangzhou, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Key Lab of Non-Coding RNA Basic and Clinical Translational Research, Yangzhou University, Yangzhou, China
| | - Hao Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China.
| | - Chun Pan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China.
| |
Collapse
|
2
|
Xie T, Zhang Z, Feng M, Kong L. Current study on Pyrroloquinoline quinone (PQQ) therapeutic role in neurodegenerative diseases. Mol Biol Rep 2025; 52:397. [PMID: 40234255 DOI: 10.1007/s11033-025-10491-6] [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: 12/10/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Pyrroloquinoline quinone (PQQ) is a naturally occurring redox-active compound with potent antioxidant, mitochondrial-enhancing, and neuroprotective properties. Originally identified as a cofactor in bacterial enzymes, PQQ has garnered significant interest for its potential therapeutic role in neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). It has reported that PQQ exerts its effects through several key molecular mechanisms, including the activation of antioxidant pathways via Nrf2/ARE signaling, enhancement of mitochondrial biogenesis and function through AMPK/PGC-1α, and the regulation of inflammatory processes through NF-κB inhibition. By improving cellular energy metabolism, reducing oxidative stress, and promoting neuronal survival, PQQ offers a multifaceted approach to counteracting the pathophysiological factors underlying neurodegeneration. Our review focusing on current study of PQQ on its enhancing neuroplasticity, and protecting neurons from damage induced by oxidative stress, mitochondrial dysfunction, and inflammation. Further we reviewed the significant signaling pathways that involved PQQ neuroprotective mechanisms, positioning it as a novel candidate for future therapeutic strategies targeting these debilitating conditions.
Collapse
Affiliation(s)
- Tao Xie
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
- Medical College, Yan'an University, Yan'an City, Shaanxi, China
| | - Zhen Zhang
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Mingzhe Feng
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingbo Kong
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
3
|
Shi J, Dai L, Gu J, Liu L, Chen Y, Jiang Y, Yu Y. Luteolin alleviates olfactory dysfunction in eosinophilic chronic rhinosinusitis through modulation of the TLR4/NF-κB signaling pathway. Int Immunopharmacol 2025; 148:114189. [PMID: 39892170 DOI: 10.1016/j.intimp.2025.114189] [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: 11/05/2024] [Revised: 01/07/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVES Eosinophilic chronic rhinosinusitis (ECRS) is characterized by early, severe olfactory dysfunction and a high recurrence rate, with inadequate treatments. This article aims to elucidate the potential mechanisms by which luteolin may treat olfactory dysfunction in the context of ECRS. METHODS Thirty C57 BL/6 mice were randomly assigned to five groups: a control group and four experimental groups (ECRS, ECRS + Luteolin (Low, Medium, High Dose)). We conducted RNA sequencing, behavioral testing, ELISA, PCR, HE staining, Western blot analysis, and immunofluorescence staining. Additionally, human olfactory epithelial Cells (HOEPCs) were treated with Luteolin, TAK-242, QNZ, and Luteolin + LPS to investigate the underlying mechanisms. RESULTS Luteolin significantly reduced IL-4, IL-5, and IL-13 levels in the nasal lavage fluid (NLF) of ECRS mice, improving olfactory function and restoring OMP+ mature and Gap43+ immature olfactory sensory neurons (OSNs). RNA sequencing revealed the involvement of the TLR4/NF-κB pathway in ECRS-related olfactory dysfunction and luteolin's therapeutic effects. Luteolin reduced TLR4+ cells, P65 nuclear translocation, and decreased protein levels of IL-1β, TLR4, MyD88, p-P65, P65, and p-P38. The treatment also lowered OSNs cell apoptosis by decreasing the levels of cleaved caspase-3 and caspase-9 levels, and increasing Bcl-2 protein. Antioxidant enzymes SOD, CAT, and GSH-Px were elevated, while MDA levels decreased. In ECRS HOEPCs, luteolin's anti-apoptotic effects on OSNs were reversed by LPS-induced TLR4/NF-κB activation. CONCLUSIONS Luteolin ameliorates olfactory dysfunction associated with ECRS by modulating the TLR4/NF-κB signaling pathway. This modulation results in a reduction of TH2-type inflammation, oxidative stress, and apoptosis in OSNs.
Collapse
Affiliation(s)
- Jiali Shi
- Department of Otorhinolaryngology, The First Affiliated Hospital of Soochow University Suzhou Jiangsu PR China; Department of Otorhinolaryngology, Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University. Shanghai PR China.
| | - Li Dai
- Department of Otorhinolaryngology, Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University. Shanghai PR China.
| | - Jun Gu
- Department of Otorhinolaryngology, Changshu Second People's Hospital Jiangsu PR China.
| | - Lu Liu
- Center for Drug Safety Evaluation and Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai PR China.
| | - Yunhua Chen
- Department of Otorhinolaryngology, Changshu Second People's Hospital Jiangsu PR China.
| | - Yiming Jiang
- Department of Otorhinolaryngology, Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University. Shanghai PR China.
| | - Yafeng Yu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Soochow University Suzhou Jiangsu PR China.
| |
Collapse
|
4
|
Holt R, Yahyavi SK, Kooij I, Poulsen NN, Juul A, Jørgensen N, Blomberg Jensen M. Effects of vitamin D on sex steroids, luteinizing hormone, and testosterone to luteinizing hormone ratio in 307 infertile men. Andrology 2024; 12:553-560. [PMID: 37555466 DOI: 10.1111/andr.13505] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/07/2023] [Accepted: 07/29/2023] [Indexed: 08/10/2023]
Abstract
OBJECTIVE Vitamin D status has been associated with sex steroid production. The question is whether vitamin D supplementation has an impact on sex steroid production in infertile men with vitamin D insufficiency? DESIGN A single-center, double-blinded, randomized clinical trial. Differences in sex steroids and reproductive hormones were predefined secondary outcomes, vitamin D status at baseline was a predefined subgroup and the primary outcome was differences in semen quality. METHODS A total of 307 infertile men were included and randomized 1:1 to active or placebo treatment for 150 days. Men in the active group initially received an oral bolus of 300,000 IU cholecalciferol, followed by daily supplementation with 1400 IU cholecalciferol and 500 mg calcium. RESULTS After intervention, no differences were found in serum concentrations of sex steroids, luteinizing hormone, testosterone/luteinizing hormone ratio or SHBG between the vitamin D and placebo group. However, in a predefined subgroup analysis of men with serum 25OHD ≤ 50 nmol/L, men treated with vitamin D had a significantly higher testosterone/luteinizing hormone ratio [4.2 (3.8-4.4) vs. 3.7 (3.4-4.0); p = 0.033] compared with placebo treatment. In men with vitamin D deficiency, the difference between groups was larger but not significant due to few men with serum 25OHD < 25 nmol/L. CONCLUSION Vitamin D + calcium supplementation did not alter sex steroid production in infertile men. However, vitamin D insufficient men treated with vitamin D supplementation had a significantly higher testosterone/LH ratio compared with placebo-treated men, suggesting that optimal Leydig cell function are dependent on adequate vitamin D status.
Collapse
Affiliation(s)
- Rune Holt
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Sam Kafai Yahyavi
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Ireen Kooij
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Nadia Nicholine Poulsen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| |
Collapse
|
5
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Antiaging Strategies and Remedies: A Landscape of Research Progress and Promise. ACS Chem Neurosci 2024; 15:408-446. [PMID: 38214973 PMCID: PMC10853939 DOI: 10.1021/acschemneuro.3c00532] [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: 08/12/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Aging is typified by a gradual loss of physiological fitness and accumulation of cellular damage, leading to deteriorated functions and enhanced vulnerability to diseases. Antiaging research has a long history throughout civilization, with many efforts put forth to understand and prevent the effects of aging. Multiple strategies aiming to promote healthy aging and extend the lifespan have been developed including lifestyle adjustments, medical treatments, and social programs. A multitude of antiaging medicines and remedies have also been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent research related to antiaging strategies and treatments. We review the recent advances and delineate trends in research headway of antiaging knowledge and practice across time, geography, and development pipelines. We further assess the state-of-the-art antiaging approaches and explore their correlations with age-related diseases. The landscape of antiaging drugs has been outlined and explored. Well-recognized and novel, currently evaluated antiaging agents have also been summarized. Finally, we review clinical applications of antiaging products with their development pipelines. The objective of this review is to summarize current knowledge on preventive strategies and treatment remedies in the field of aging, to outline challenges and evaluate growth opportunities, in order to further efforts to solve the problems that remain.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American
Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
6
|
Chen B, Chen L, Yang Z, Fu Q, Li X, Cao C. Acute Aluminum Sulfate Triggers Inflammation and Oxidative Stress, Inducing Tissue Damage in the Kidney of the Chick. Biol Trace Elem Res 2023; 201:1442-1450. [PMID: 35551605 DOI: 10.1007/s12011-022-03260-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/21/2022] [Indexed: 02/07/2023]
Abstract
In this study, a total of 20 7-day-old chicks were randomly divided into an experimental group and a control group. The experimental group was administered aluminum sulfate (Al2(SO4)3) once by gavage, and the control group was sacrificed after 24 h of fasting with distilled water. Serum and kidney tissue samples from both groups were collected and compared using hematoxylin-eosin staining (H&E) and microscopy. The Paller scores increased (p < 0.01) for biochemical kidney function, redox-related indicators, and mRNA expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) downstream related genes. The results showed that in the kidneys of the experimental group, renal tubular epithelial cells appeared to swell, and there was necrosis and shedding; the blood urea nitrogen (BUN) and uric acid (UA) decreased, serum creatinine (CREA) increased; nitric oxide (NO), glutathione (GSH), and malondialdehyde (MDA) contents increased; NO synthase (NOS), glutathione peroxidase (GSH-PX), and superoxide dismutase (SOD) enzyme activities increased; tumor necrosis factor alpha (TNF-α), tumor necrosis factor receptor 1 (TNF-R1), tumor necrosis factor receptor 2 (TNF -R2), cyclooxygenase-2 (COX-2), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and heme oxygenase-1 (HO-1) mRNA expression levels increased (p < 0.05 or p < 0.01); Nrf2, glutathione S-transferase A3 (GSTA3), glutathione-S-transferase mu-1 (GSTM1), glutathione synthetase (GSS), glutamate cysteine ligase (GCLC and GCLM), quinone oxidoreductase 1 (NQO1), and Kelch-like ECH-associated protein 1 (Keap1) mRNA expression levels decreased (p < 0.05 or p < 0.01) compared to the control group. Acute aluminum poisoning can cause obvious pathological changes in the structure of the kidney tissue of the chick, resulting in damage to the kidney function, as well as triggering inflammation and oxidative stress in the kidney.
Collapse
Affiliation(s)
- Bo Chen
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China
| | - Lina Chen
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China
| | - Zhiqing Yang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China
| | - Qiang Fu
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China
| | - Xinran Li
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China.
- Foshan University Veterinary Teaching Hospital, Foshan, Guangdong, 528231, People's Republic of China.
| | - Changyu Cao
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, 528231, People's Republic of China.
| |
Collapse
|
7
|
Kou Y, Rong X, Tang R, Zhang Y, Yang P, Liu H, Ma W, Li M. Eldecalcitol prevented OVX-induced osteoporosis through inhibiting BMSCs senescence by regulating the SIRT1-Nrf2 signal. Front Pharmacol 2023; 14:1067085. [PMID: 36937895 PMCID: PMC10020367 DOI: 10.3389/fphar.2023.1067085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background: Aging and oxidative stress are considered to be the proximal culprits of postmenopausal osteoporosis. Eldecalcitol (ED-71), a new active vitamin D derivative, has shown a good therapeutic effect on different types of osteoporosis, but the mechanism is unclear. This study focused on exploring whether ED-71 could prevent bone loss in postmenopausal osteoporosis by regulating the cell senescence of bone mesenchymal stem cells (BMSCs), and explaining its specific mechanism of action. Materials and methods: An ovariectomized (OVX) rat model was established and 30 ng/kg ED-71 was administered orally once a day. The weight of rats was recorded regularly. Micro-computed tomography (CT) and histochemical staining were used to evaluate bone mass, histological parameters, and aging-related factors. Rat bone mesenchymal stem cells were extracted and cultivated in vitro. Aging cells were marked with senescence-associated β-gal (SA-β-gal) dyeing. The mRNA and protein levels of aging-related factors and SIRT1-Nrf2 signal were detected by RT-PCR, Western blot, and immunofluorescence staining. The reactive oxygen species (ROS) levels were detected by DCFH-DA staining. Results: Compared with the Sham group, the bone volume of the ovariectomized group rats decreased while their weight increased significantly. ED-71 prevented bone loss and inhibited weight gain in ovariectomized rats. More importantly, although the expression of aging-related factors in the bone tissue increased in the ovariectomized group, the addition of ED-71 reversed changes in these factors. After extracting and in vitro culturing bone mesenchymal stem cells, the proportion of aging bone mesenchymal stem cells was higher in the ovariectomized group than in the Sham group, accompanied by a significant decrease in the osteogenic capacity. ED-71 significantly improved the bone mesenchymal stem cells senescence caused by ovariectomized. In addition, ED-71 increased the expression of SIRT1 and Nrf2 in ovariectomized rat bone mesenchymal stem cells. Inhibition of SIRT1 or Nrf2 decreased the inhibitory effect of ED-71 on bone mesenchymal stem cells senescence. ED-71 also showed a suppression effect on the reactive oxygen species level in bone mesenchymal stem cells. Conclusion: Our results demonstrated that ED-71 could inhibit the cell senescence of bone mesenchymal stem cells in ovariectomized rats by regulating the SIRT1-Nrf2 signal, thereby preventing bone loss caused by osteoporosis.
Collapse
Affiliation(s)
- Yuying Kou
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Xing Rong
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Rong Tang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Yuan Zhang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Panpan Yang
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Hongrui Liu
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
| | - Wanli Ma
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
- Department of Traumatic Orthopedics, The Second Hospital of Shandong University, Jinan, China
- *Correspondence: Wanli Ma, ; Minqi Li,
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, China
- *Correspondence: Wanli Ma, ; Minqi Li,
| |
Collapse
|
8
|
Miao D, Goltzman D. Mechanisms of action of vitamin D in delaying aging and preventing disease by inhibiting oxidative stress. VITAMINS AND HORMONES 2022; 121:293-318. [PMID: 36707138 DOI: 10.1016/bs.vh.2022.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although several recent studies have shown that vitamin D supplementation beneficially decreases oxidative stress parameters, there is no consensus on this subject in humans. Thus the role of vitamin D supplementation has recently become a controversial topic because large intervention studies in humans have not shown significant benefits. These studies have indicated that supplementation with precursor forms of active vitamin D has no effect on all-cause mortality, cannot reduce the fracture risk of the elderly, cannot reduce the incidence of cancer or cardiovascular disease in the elderly, and cannot significantly reduce the incidence risk of diabetes in the elderly. However, a link between several age-related diseases and enhanced oxidative stress has been found in mice with insufficient or deficient 1,25-dihydroxyvitamin D (1,25(OH)2D), the active form of vitamin D, which indicates that reduced active vitamin D accelerates aging and age-related diseases by increasing oxidative stress. Furthermore, supplementation of exogenous 1,25(OH)2D3, or antioxidants, could dramatically postpone aging, prevent osteoporosis and spontaneous tumor development induced by 1,25(OH)2D insufficiency or deficiency, by inhibiting oxidative stress. Mechanistically, the antioxidative effects of 1,25(OH)2D3 are carried out via the vitamin D receptor (VDR) by activation of the Nrf2 oxidative stress response pathway though transcriptional or posttranscriptional activation of Nrf2 or transcriptional upregulation of Sirt1 and Bmi1 expression. Whether discrepancies between studies in humans and in mice reflect the different forms of vitamin D examined remains to be determined.
Collapse
Affiliation(s)
| | - David Goltzman
- McGill University Health Centre and McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Liu Y, Li Y, Niu J, Liu H, Jiao N, Huang L, Jiang S, Yan L, Yang W. Effects of Dietary Macleaya cordata Extract Containing Isoquinoline Alkaloids Supplementation as an Alternative to Antibiotics in the Diets on Growth Performance and Liver Health of Broiler Chickens. Front Vet Sci 2022; 9:950174. [PMID: 35968000 PMCID: PMC9363708 DOI: 10.3389/fvets.2022.950174] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 06/21/2022] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate the effects of dietary supplementation with Macleaya cordata extract (MCE) containing protopine and allotypotopine on the growth performance and liver health in broiler chickens. A total of 486 1-day-old male AA broiler chickens were randomly assigned to the following three groups: (1) control (CON) group, broiler chickens fed a basal diet; (2) AGP group (positive control), broiler chickens fed a basal diet supplemented with 50 mg/kg aureomycin; (3) MCE group, broiler chickens fed a basal diet supplemented with 0.6 mg/kg MCE including 0.4 mg/kg protopine and 0.2 mg/kg allotypotopine. The results showed that the MCE group had significantly higher final body weight and average daily gain from d 0 to 42 than the other groups (p < 0.05), and groups MCE and AGP both had significantly lower feed-to-gain ratio from d 0 to 42 than the CON group (p < 0.05). Serum total protein, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, total cholesterol, glucose, immunoglobulin A, immunoglobulin M, and complements (C3, C4) concentrations in the MCE group were significantly higher than in the CON group (p < 0.05). Dietary MCE or aureomycin supplementation significantly reduced the hepatic contents of 8-hydroxy-2'-deoxyguanosine, malondialdehyde, interleukin (IL)-1β, IL-6, NLRs family pyrin domain containing 3 (NLRP3), and caspase-1 in the liver (p < 0.05). Moreover, MCE or aureomycin supplementation significantly inhibited mRNA expressions of Toll-like receptor 4, myeloid differentiation factor 88, nuclear factor-κB, and NLRP3, as well as the expression ratio of Bax to Bcl-2 mRNA (p < 0.05). Therefore, our study suggested that dietary supplementation with 0.6 mg/kg MCE containing protopine and allocryptopine improved growth performance and benefited liver health in broiler chickens possibly through inhibiting caspase-1-induced pyroptosis by inactivating TLR4/MyD88/NF-κB/NLRP3 signaling pathway, and provided support for the application of MCE containing protopine and allocryptopine as an alternative to antibiotics in the feed industry.
Collapse
Affiliation(s)
- Yang Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Yang Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
- *Correspondence: Yang Li
| | - Jiaxing Niu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Hua Liu
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Ning Jiao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Libo Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Shuzhen Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Lei Yan
- Shandong New Hope Liuhe Group Co., Ltd., Qingdao, China
| | - Weiren Yang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
- Weiren Yang
| |
Collapse
|
10
|
Chen Y, Hamidu S, Yang X, Yan Y, Wang Q, Li L, Oduro PK, Li Y. Dietary Supplements and Natural Products: An Update on Their Clinical Effectiveness and Molecular Mechanisms of Action During Accelerated Biological Aging. Front Genet 2022; 13:880421. [PMID: 35571015 PMCID: PMC9096086 DOI: 10.3389/fgene.2022.880421] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
Accelerated biological aging, which involves the gradual decline of organ or tissue functions and the distortion of physiological processes, underlies several human diseases. Away from the earlier free radical concept, telomere attrition, cellular senescence, proteostasis loss, mitochondrial dysfunction, stem cell exhaustion, and epigenetic and genomic alterations have emerged as biological hallmarks of aging. Moreover, nutrient-sensing metabolic pathways are critical to an organism's ability to sense and respond to nutrient levels. Pharmaceutical, genetic, and nutritional interventions reverting physiological declines by targeting nutrient-sensing metabolic pathways can promote healthy aging and increase lifespan. On this basis, biological aging hallmarks and nutrient-sensing dependent and independent pathways represent evolving drug targets for many age-linked diseases. Here, we discuss and update the scientific community on contemporary advances in how dietary supplements and natural products beneficially revert accelerated biological aging processes to retrograde human aging and age-dependent human diseases, both from the clinical and preclinical studies point-of-view. Overall, our review suggests that dietary/natural products increase healthspan-rather than lifespan-effectively minimizing the period of frailty at the end of life. However, real-world setting clinical trials and basic studies on dietary supplements and natural products are further required to decisively demonstrate whether dietary/natural products could promote human lifespan.
Collapse
Affiliation(s)
- Ye Chen
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sherif Hamidu
- Clinical Pathology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Xintong Yang
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiqi Yan
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qilong Wang
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Patrick Kwabena Oduro
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Clinical Pathology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Yuhong Li
- State Key Laboratory of Pharmacology of Modern Chinese Medicine, Department of Pharmacology and Toxicology, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
11
|
Boretti A. PQQ Supplementation and SARS-CoV-2 Spike Protein-Induced Heart Inflammation. Nat Prod Commun 2022; 17:1934578X221080929. [PMID: 35287384 PMCID: PMC8905048 DOI: 10.1177/1934578x221080929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/31/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 spike protein-induced heart inflammation may originate from either COVID-19 infection or the administration of COVID-19 mRNA vaccines. As pyrroloquinoline quinone (PQQ) is a scavenger of free radicals, redox cofactor, and antioxidant which supports cognitive and mitochondrial functions, supplementation with PQQ could have a positive effect to reduce heart inflammation after COVID-19 mRNA vaccines. However, there is no evidence yet for this opportunity in the literature. Cellular and animal model results are missing. Similarly, no clinical trials have been conducted. While it is recommended to measure the levels of the cardiac biomarkers before and after COVID-19 vaccination, no recommendation can be made about supplementation with PQQ, which, however, we note has no contraindication.
Collapse
Affiliation(s)
- Alberto Boretti
- Independent Scientist, Johnsonville Road, Johnsonville, Wellington 6037, New Zealand
| |
Collapse
|
12
|
Long C, Wang Z, Guo Y, Sheng X, Xing K, Ni H, Wang X, Xiao L, Qi X. Research Note: Dietary supplementation with pyrroloquinoline quinone disodium (PQQ.Na 2) improves oxidative status and semen quality in aging layer breeder roosters. Poult Sci 2022; 101:101812. [PMID: 35325831 PMCID: PMC8942821 DOI: 10.1016/j.psj.2022.101812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022] Open
Abstract
As the antioxidant capacity of sperm declines with age in roosters, the objective of the present study was to determine the effects of different levels of pyrroloquinoline quinone disodium (PQQ.Na2) on antioxidative and sperm quality parameters of aging layer breeder roosters. A total of ninety-six 63-wk-old Jinghong No. 1 layer breeder roosters were randomly assigned to 4 treatments (0, 0.5, 1, 2 mg/kg PQQ.Na2) for 6 wk. Antioxidant activity and semen parameters were assessed biweekly. The dietary administration of PQQ.Na2 significantly increased semen quality (semen volume, sperm motility, straightness, progressive motility, curvilinear velocity, straight-line velocity, and amplitude of lateral head displacement) and antioxidant capacity (T-SOD, GSH-Px, hydroxyl radical scavenging ability, and/or superoxide scavenging capacity) in seminal plasma in aging layer breeder roosters. Whereas, PQQ.Na2 supplementations significantly decreased malondialdehyde (MDA) concentration in seminal plasma in aging layer breeder roosters. Supplementation with 1 mg/kg dietary PQQ.Na2 as an antioxidant supplement could increase sperm quality and antioxidant activity of aging layer breeder roosters, while a higher dose (2 mg/kg) did not result in further increment.
Collapse
Affiliation(s)
- Cheng Long
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Ziteng Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Yong Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Kai Xing
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Heming Ni
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China.
| |
Collapse
|
13
|
Zhao L, Mao Z, Mou D, Huang L, Yang M, Ding D, Yan H, Fang Z, Che L, Zhuo Y, Jiang X, Xu S, Lin Y, Li J, Huang C, Zou Y, Li L, Wu D, Feng B. Maternal cholecalciferol supplementation during gestation improves antioxidant capacities in gilts and piglets. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1961616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Lianpeng Zhao
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhengyu Mao
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Daolin Mou
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Long Huang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Min Yang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Dajing Ding
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Jiang
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yan Lin
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jian Li
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - De Wu
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bin Feng
- Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|