1
|
Delen O, Uz YH, Yuksel C, Ersoy O, Kizilay G. Gallic acid mitigates lipopolysaccharide-induced testicular inflammation via regulation of the NF-κB and PK2/PKR1 pathway. J Mol Histol 2025; 56:71. [PMID: 39825958 DOI: 10.1007/s10735-024-10349-4] [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] [Received: 11/22/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
Genital tract infections are common causes of male infertility, and most of diagnosed men are asymptomatic. This study examined the effect of gallic acid (GA) against lipopolysaccharide (LPS)-induced testicular inflammation. Thirty-two Spraque Dawley, 2.5-3 month-old male rats were separated into four groups (n = 8). Control group; saline at 3 ml/kg, and in the GA group; GA was dissolved in saline, by gavage at 100 mg/kg for 14 days. LPS group; LPS 5 mg/kg as a single dose was given intraperitoneal on the 11th day. LPS + GA group; GA was given for 14 days and LPS 5 mg/kg on the 11th day. After 72 h of LPS injection, all samples were collected. Semen analysis, biochemical assays, histological evaluations, and immunohistochemical or Western blot analyses for nuclear factor-kappa B (NF-κB) and Prokineticin 2/prokineticin receptor 1(PK2/PKR1) pathways were performed. There was a significant decrease in body and testicular weight, sperm parameters, serum testosterone level, mean seminiferous tubule diameter, germinal epithelial thickness, and Johnsen score in the LPS group compared to control and GA groups. However, a significant increase was found in interstitial space width, percentage of abnormal sperm, NF-κB and PK2 immunoreactivities, and expression of PK2 and PKR1 proteins. In the LPS + GA group, GA administration was observed to significantly prevent these adverse effects. In conclusion, the inhibitory effects of GA on the NF-κB and PK2/PKR1 pathways not only suppressed the inflammatory response but also restored impaired sperm parameters and testicular structure. These findings indicate GA's potential for treating testicular inflammation and protecting male reproductive health.
Collapse
Affiliation(s)
- Ozlem Delen
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, 22030, Turkey.
| | - Yesim Hulya Uz
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, 22030, Turkey
| | - Cengiz Yuksel
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, 22030, Turkey
| | - Onur Ersoy
- Department of Pathology Laboratory Techniques, Vocational School of Health Services, Trakya University, Edirne, Turkey
| | - Gulnur Kizilay
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, 22030, Turkey
| |
Collapse
|
2
|
Luo YW, Zhu XL, Yang ZM, Zhou JH, Tao T, Chen BH, Qin SL, Liu BL, Hu W. Adrenomedullin gene delivery rescues estrogen production in Leydig cells via the inhibition of TGF-β1/Smads signaling pathway. Reprod Toxicol 2025; 132:108834. [PMID: 39793742 DOI: 10.1016/j.reprotox.2025.108834] [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: 11/13/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Our previous findings demonstrated that adrenomedullin (ADM) protects against the reduction in testosterone production and apoptosis of Leydig cells both in vitro and in vivo. In this study, we investigated whether ADM could preserve estrogen production in Leydig cells by suppressing the transforming growth factor-β1 (TGF-β1) / Smads signaling pathway. Leydig cells were treated with lipopolysaccharide (LPS) and recombinant adenovirus ADM (Ad-ADM), an adeno-associated viral vector expressing ADM. Cell viability and cytochrome P450 aromatase (P450arom) activity were assessed. Estrogen, testosterone, and TGF-β1 concentrations in the culture medium were measured. Additionally, the gene expression and protein levels of CYP19, TGF-β1, and Smads were evaluated. The results indicated that Ad-ADM mitigated the reductions in Leydig cell viability and testosterone production, counteracted the decreases in P450arom activity, and restored CYP19 gene expression and protein levels in LPS-treated cells. Moreover, Ad-ADM reduced the elevated gene expression and protein levels of Smads and TGF-β1 induced by LPS. Based on these findings, we propose that ADM safeguards estrogen production in Leydig cells by inhibiting the TGF-β1/Smads signaling pathway.
Collapse
Affiliation(s)
- You-Wen Luo
- Department of Ultrasound Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Xia-Lian Zhu
- Department of Nuclear Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, China
| | - Zhi-Min Yang
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Jian-Hua Zhou
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Tong Tao
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Bing-Hai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Song-Lin Qin
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Bo-Long Liu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Wei Hu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China.
| |
Collapse
|
3
|
Fomichova O, Oliveira PF, Bernardino RL. Exploring the interplay between inflammation and male fertility. FEBS J 2024. [PMID: 39702986 DOI: 10.1111/febs.17366] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Male fertility results from a complex interplay of physiological, environmental, and genetic factors. It is conditioned by the properly developed anatomy of the reproductive system, hormonal regulation balance, and the interplay between different cell populations that sustain an appropriate and functional environment in the testes. Unfortunately, the mechanisms sustaining male fertility are not flawless and their perturbation can lead to infertility. Inflammation is one of the factors that contribute to male infertility. In the testes, it can be brought on by varicocele, obesity, gonadal infections, leukocytospermia, physical obstructions or traumas, and consumption of toxic substances. As a result of prolonged or untreated inflammation, the testicular resident cells that sustain spermatogenesis can suffer DNA damage, lipid and protein oxidation, and mitochondrial dysfunction consequently leading to loss of function in affected Sertoli cells (SCs) and Leydig cells (LCs), and the formation of morphologically abnormal dysfunctional sperm cells that lay in the basis of male infertility and subfertility. This is due mainly to the production and secretion of pro-inflammatory mediators, including cytokines, chemokines, and reactive oxygen species (ROS) by local immune cells (macrophages, lymphocytes T, mast cells) and tissue-specific cells [SCs, LCs, peritubular myoid cells (PMCs) and germ cells (GCs)]. Depending on the location, duration, and intensity of inflammation, these mediators can exert their toxic effect on different elements of the testes. In this review, we discuss the most prevalent inflammatory factors that negatively affect male fertility and describe the different ways inflammation can impair male reproductive function.
Collapse
Affiliation(s)
- Oleksandra Fomichova
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, Portugal
| | - Raquel L Bernardino
- UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Portugal
| |
Collapse
|
4
|
Chen Z, Jiang M, Mo L, Zhou C, Huang H, Ma C, Wang Z, Fan Y, Chen Z, Fang B, Liu Y. A natural agent, 5-deoxycajanin, mitigates estrogen-deficiency bone loss via modulating osteoclast-osteoblast homeostasis. Int Immunopharmacol 2024; 141:112906. [PMID: 39173403 DOI: 10.1016/j.intimp.2024.112906] [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: 06/05/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Hyperactive osteoclasts and hypoactive osteoblasts usually result in osteolytic conditions such as estrogen-deficiency bone loss. Few natural compounds that both attenuating bone resorption and enhancing bone formation could exert effects on this imbalance. 5-Deoxycajanin (5-D), an isoflavonoid extracted from Cajan leaf with estrogen-like properties, were found to have beneficial pharmacological effects on rebalancing the activities of osteoclasts and osteoblasts. This study revealed that 5-D at the same concentration could inhibit osteoclastogenesis of BMMs and promoted osteoblast differentiation of BMSCs. 5-D not only attenuated the fluorescent formation of RANKL-induced F-actin belts and NFATc1, but also activated ALP and RUNX2 expressions. As to downstream factor expressions, 5-D could block osteoclast-specific genes and proteins including NFATc1 and CTSK, while increased osteogenic genes and proteins including OPG and OCN, as confirmed by Real-time PCR and Western Blotting. Additionally, the network pharmacology and molecular docking identified the involvement of 5-D in the MIF and MAPK signaling pathways and the stable binding between 5-D and MAPK2K1. Further Western blot studies showed that 5-D decreased the phosphorylation of p38 and ERK in osteoclasts, but promoted these phosphorylations in osteoblasts. In a female C57BL/6J mouse model of estrogen deficiency-induced bone loss, 5-D demonstrated efficacy in enhancing BMD through attenuating osteoclast activities and promoting osteogenesis. These results underscore the potential application of 5-D on treating osteolysis resulting from hyperactive osteoclasts and hypoactive osteoblasts, shedding light on modulating osteoclast-osteoblast homeostasis.
Collapse
Affiliation(s)
- Zhiwen Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengyu Jiang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liang Mo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chi Zhou
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoran Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Yangjiang Hospital of Traditional Chinese Medicine, Yangjiang, China
| | - Chao Ma
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhangzheng Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinuo Fan
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenqiu Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Bin Fang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yuhao Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
5
|
Nájera-Martínez M, Lara-Vega I, Avilez-Alvarado J, Pagadala NS, Dzul-Caamal R, Domínguez-López ML, Tuszynski J, Vega-López A. The Generation of ROS by Exposure to Trihalomethanes Promotes the IκBα/NF-κB/p65 Complex Dissociation in Human Lung Fibroblast. Biomedicines 2024; 12:2399. [PMID: 39457711 PMCID: PMC11505202 DOI: 10.3390/biomedicines12102399] [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: 07/06/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Disinfection by-products used to obtain drinking water, including halomethanes (HMs) such as CH2Cl2, CHCl3, and BrCHCl2, induce cytotoxicity and hyperproliferation in human lung fibroblasts (MRC-5). Enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) modulate these damages through their biotransformation processes, potentially generating toxic metabolites. However, the role of the oxidative stress response in cellular hyperproliferation, modulated by nuclear factor-kappa B (NF-κB), remains unclear. Methods: In this study, MRC-5 cells were treated with these compounds to evaluate reactive oxygen species (ROS) production, lipid peroxidation, phospho-NF-κB/p65 (Ser536) levels, and the activities of SOD, CAT, and GPx. Additionally, the interactions between HMs and ROS with the IκBα/NF-κB/p65 complex were analyzed using molecular docking. Results: Correlation analysis among biomarkers revealed positive relationships between pro-oxidant damage and antioxidant responses, particularly in cells treated with CH2Cl2 and BrCHCl2. Conversely, negative relationships were observed between ROS levels and NF-κB/p65 levels in cells treated with CH2Cl2 and CHCl3. The estimated relative free energy of binding using thermodynamic integration with the p65 subunit of NF-κB was -3.3 kcal/mol for BrCHCl2, -3.5 kcal/mol for both CHCl3 and O2•, and -3.6 kcal/mol for H2O2. Conclusions: Chloride and bromide atoms were found in close contact with IPT domain residues, particularly in the RHD region involved in DNA binding. Ser281 is located within this domain, facilitating the phosphorylation of this protein. Similarly, both ROS interacted with the IPT domain in the RHD region, with H2O2 forming a side-chain oxygen interaction with Leu280 adjacent to the phosphorylation site of p65. However, the negative correlation between ROS and phospho-NF-κB/p65 suggests that steric hindrance by ROS on the C-terminal domain of NF-κB/p65 may play a role in the antioxidant response.
Collapse
Affiliation(s)
- Minerva Nájera-Martínez
- Laboratorio de Toxicología Ambiental, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, Mexico City 07738, Mexico; (M.N.-M.); (I.L.-V.)
| | - Israel Lara-Vega
- Laboratorio de Toxicología Ambiental, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, Mexico City 07738, Mexico; (M.N.-M.); (I.L.-V.)
| | - Jhonatan Avilez-Alvarado
- Laboratorio de Visión Artificial, Unidad Culhuacán, Escuela Superior de Ingeniería Mecánica y Eléctrica, Instituto Politécnico Nacional, Av. Santa Ana 1000, San Francisco Culhuacán CTM V, Mexico City 04440, Mexico;
| | | | - Ricardo Dzul-Caamal
- Instituto EPOMEX, Universidad Autónoma de Campeche, Av. Héroe de Nacozari No. 480, Campeche 24070, Mexico;
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica I, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Carpio y Plan de Ayala s/n, Casco de Santo Tomás, Mexico City 11340, Mexico;
| | - Jack Tuszynski
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| | - Armando Vega-López
- Laboratorio de Toxicología Ambiental, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, Mexico City 07738, Mexico; (M.N.-M.); (I.L.-V.)
| |
Collapse
|
6
|
Zhong Y, Liang B, Zhang X, Li J, Zeng D, Huang T, Wu J. NF-κB affected the serum levels of TNF-α and IL-1β via activation of the MAPK/NF-κB signaling pathway in rat model of acute pulmonary microthromboembolism. Pulm Circ 2024; 14:e12357. [PMID: 38584678 PMCID: PMC10995479 DOI: 10.1002/pul2.12357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024] Open
Abstract
Pulmonary thromboembolism caused by thrombi blocking major pulmonary artery and its branches, is a frequently encountered phenomenon and an important cause of high morbidity and mortality in lung diseases and may develop into persistent pulmonary hypertension (PH). Nuclear factor-κB (NF-κB) signaling pathway had been reported participated in the formation and development of PH by promoting inflammatory response. The aim of this study was to investigate the effects of NF-κB activation on the serum levels of tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β) in acute pulmonary microthromboembolism (APMTE) rats. Rats were randomized into five groups. APMTE group received jugular vein injection of autologous thrombus, while control group rats received normal saline injection. Pulmonary hemodynamic parameters were measured through ECHO-guided transthoracic puncture. Pulmonary vascular morphological changes were analyzed by HE. The expression changes of NF-κB and serum TNF-α、IL-1β levels were detected by enzyme-linked immunosorbent assay. Protein expression of the MAPK/NF-κB signaling pathway including p-IκBα, p-p38 MAPK, p-NF-κB p65, IκBα, p38 MAPK, and NF-κB p65 was determined using western blot analysis. Compared with control group, the expression of NF-κB in lung tissue and the levels of serum TNF-α and IL-1β rats were higher, a significant reduction in IκBα and elevation in the phosphorylation of IκBα, p38 MAPK, and NF-κB p65 were found in APMTE group rats. And UK administration reversed the APMTE-induced increase in TNF-α, IL-1β, p-IκBα, p-MAPK, and p-NF-κB protein. Furthermore, the levels of NF-κB, TNF-α, and IL-1β were positively correlated with mean pulmonary artery. And the levels of TNF-α and IL-1β were positively correlated with NF-κB. These findings suggest that the activation of MAPK/NF-κB pathway as a critical driver of increasing TNF-α and IL-1β level in APMTE rats and UK exerted protective effects against APMTE-induced PH may be related to the downregulation of the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanfen Zhong
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Binbin Liang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Xiaofeng Zhang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jingtao Li
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Decai Zeng
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Tongtong Huang
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ji Wu
- Department of Ultrasonic MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
7
|
Xing D, Jin Y, Jin B. A narrative review on inflammaging and late-onset hypogonadism. Front Endocrinol (Lausanne) 2024; 15:1291389. [PMID: 38298378 PMCID: PMC10827931 DOI: 10.3389/fendo.2024.1291389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
The increasing life expectancy observed in recent years has resulted in a higher prevalence of late-onset hypogonadism (LOH) in older men. LOH is characterized by the decline in testosterone levels and can have significant impacts on physical and mental health. While the underlying causes of LOH are not fully understood, there is a growing interest in exploring the role of inflammaging in its development. Inflammaging is a concept that describes the chronic, low-grade, systemic inflammation that occurs as a result of aging. This inflammatory state has been implicated in the development of various age-related diseases. Several cellular and molecular mechanisms have been identified as contributors to inflammaging, including immune senescence, cellular senescence, autophagy defects, and mitochondrial dysfunction. Despite the extensive research on inflammaging, its relationship with LOH has not yet been thoroughly reviewed in the literature. To address this gap, we aim to review the latest findings related to inflammaging and its impact on the development of LOH. Additionally, we will explore interventions that target inflammaging as potential treatments for LOH.
Collapse
Affiliation(s)
- Dong Xing
- Medical College of Southeast University, Nanjing, Jiangsu, China
| | - Yihan Jin
- Reproductive Medicine Center, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Baofang Jin
- Andrology Department of Integrative Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Luo YW, Zhu XL, Li MY, Zhou JH, Yang ZM, Tong T, Chen BH, Qin SL, Liu BL, Hu W. Anti-apoptotic effect of adrenomedullin gene delivery on Leydig cells by suppressing TGF-β1 via the Hippo signaling pathway. Reprod Toxicol 2023; 119:108418. [PMID: 37268150 DOI: 10.1016/j.reprotox.2023.108418] [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: 11/11/2022] [Revised: 05/06/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023]
Abstract
This study aims to establish whether adrenomedullin (ADM) is capable to restore the steroidogenic functions of Leydig cells by suppressing transforming growth factor-β1 (TGF-β1) through Hippo signaling. Primary Leydig cells were treated with lipopolysaccharide (LPS), an adeno-associated virus vector that expressed ADM (Ad-ADM) or sh-RNA of TGF-β1 (Ad-sh-TGF-β1). The cell viability and medium concentrations of testosterone were detected. Gene expression and protein levels were determined for steroidogenic enzymes, TGF-β1, RhoA, YAP, TAZ and TEAD1. The role of Ad-ADM in the regulation of TGF-β1 promoter was confirmed by ChIP and Co-IP. Similar to Ad-sh-TGF-β1, Ad-ADM mitigated the decline in the number of Leydig cells and plasma concentrations of testosterone by restoring the gene and protein levels of SF-1, LRH1, NUR77, StAR, P450scc, 3β-HSD, CYP17 and 17β-HSD. Similar to Ad-sh-TGF-β1, Ad-ADM not only inhibited the LPS-induced cytotoxicity and cell apoptosis but also restored the gene and protein levels of SF-1, LRH1, NUR77, StAR, P450scc, 3β-HSD, CYP17 and 17β-HSD, along with the medium concentrations of testosterone in LPS-induced Leydig cells. Like Ad-sh-TGF-β1, Ad-ADM improved LPS-induced TGF-β1 expression. In addition, Ad-ADM suppressed RhoA activation, enhanced the phosphorylation of YAP and TAZ, reduced the expression of TEAD1 which interacted with HDAC5 and then bound to TGF-β1 gene promoter in LPS-exposed Leydig cells. It is thus suspected that ADM can exert anti-apoptotic effect to restore the steroidogenic functions of Leydig cells by suppressing TGF-β1 through Hippo signaling.
Collapse
Affiliation(s)
- You-Wen Luo
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xia-Lian Zhu
- Department of Nuclear Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ming-Yong Li
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jian-Hua Zhou
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhi-Min Yang
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Tao Tong
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Bing-Hai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Song-Lin Qin
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo-Long Liu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Hu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
9
|
Davison BA, Edwards C, Cotter G, Kimmoun A, Gayat É, Latosinska A, Mischak H, Takagi K, Deniau B, Picod A, Mebazaa A. Plasma and Urinary Biomarkers Improve Prediction of Mortality through 1 Year in Intensive Care Patients: An Analysis from FROG-ICU. J Clin Med 2023; 12:jcm12093311. [PMID: 37176751 PMCID: PMC10179283 DOI: 10.3390/jcm12093311] [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: 04/14/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND This study aimed to assess the value of blood and urine biomarkers in addition to routine clinical variables in risk stratification of patients admitted to ICU. METHODS Multivariable prognostic models were developed in this post hoc analysis of the French and EuRopean Outcome ReGistry in Intensive Care Units study, a prospective observational study of patients admitted to ICUs. The study included 2087 patients consecutively admitted to the ICU who required invasive mechanical ventilation or a vasoactive agent for more than 24 h. The main outcome measures were in-ICU, in-hospital, and 1 year mortality. RESULTS Models including only SAPS II or APACHE II scores had c-indexes for in-hospital and 1 year mortality of 0.64 and 0.65, and 0.63 and 0.61, respectively. The c-indexes for a model including age and estimated glomerular filtration rate were higher at 0.69 and 0.67, respectively. Models utilizing available clinical variables increased the c-index for in-hospital and 1 year mortality to 0.80 and 0.76, respectively. The addition of biomarkers and urine proteomic markers increased c-indexes to 0.83 and 0.78. CONCLUSIONS The commonly used scores for risk stratification in ICU patients did not perform well in this study. Models including clinical variables and biomarkers had significantly higher predictive values.
Collapse
Affiliation(s)
- Beth A Davison
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Momentum Research, Inc., Durham, NC 27713, USA
| | | | - Gad Cotter
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Momentum Research, Inc., Durham, NC 27713, USA
| | - Antoine Kimmoun
- Service de Médecine Intensive et Réanimation Brabois, CHRU de Nancy, Université de Lorraine, 54511 Nancy, France
- Inserm U1116, F-CRIN INI-CRCT, 54500 Nancy, France
| | - Étienne Gayat
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Department of Anesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France
- Université Paris Cité, 75006 Paris, France
| | | | | | - Koji Takagi
- Momentum Research, Inc., Durham, NC 27713, USA
| | - Benjamin Deniau
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Department of Anesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Adrien Picod
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Department of Anesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France
| | - Alexandre Mebazaa
- Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, 75010 Paris, France
- Department of Anesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, 75010 Paris, France
| |
Collapse
|
10
|
Dou W, Xie J, Chen J, Zhou J, Xu Z, Wang Z, Zhu Q. Overexpression of adrenomedullin (ADM) alleviates the senescence of human dental pulp stem cells by regulating the miR-152/CCNA2 pathway. Cell Cycle 2023; 22:565-579. [PMID: 36310381 PMCID: PMC9928452 DOI: 10.1080/15384101.2022.2135621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The limitation of human dental pulp stem cells (DPSCs), which have potential application value in regenerative medicine, is that they are prone to age in vitro. Studies have shown adrenomedullin (ADM) is believed to promote the proliferation of human DPSCs, but whether it can also affect aging remains to be investigated. A lentivirus vector was used to construct human DPSCs overexpressing ADM. Senescence tests were carried out on cells of the 7th and 15th passage. Transcriptome analysis was conducted to analyze microRNA expression regulation changes after human DPSCs overexpressed ADM. H2O2 induced the aging model of human DPSCs, and we examined the mechanism of recovery of aging through transfection experiments with miR-152 mimic, pCDH-CCNA2, and CCNA2 siRNA. Overexpression of ADM significantly upregulated the G2/M phase ratio of human DPSCs in natural passage culture (P = 0.001) and inhibited the expression of p53 (P = 0.014), P21 WAF1 (P = 0.015), and P16 INK4A (P = 0.001). Decreased ROS accumulation was observed in human DPSCs during long-term natural passage (P = 0.022). Transcriptome analysis showed that miR-152 was significantly upregulated during human DPSC senescence (P = 0.001) and could induce cell senescence by directly targeting CCNA2. Transfection with miR-152 mimic significantly reversed the inhibitory effect of ADM overexpression on p53 (P = 0.006), P21 WAF1 (P = 0.012), and P16 INK4A (P = 0.01) proteins in human DPSCs (H2O2-induced). In contrast, pCDH-CCNA2 weakened the effect of the miR-152 mimic, thus promoting cell proliferation and antiaging. ADM-overexpressing human DPSCs promote cell cycle progression and resist cellular senescence through CCNA2 expression promotion by inhibiting miR-152.
Collapse
Affiliation(s)
- Wenxue Dou
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China
| | - Jiaye Xie
- Department of Stomatology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianan Chen
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China
| | - Jiajun Zhou
- Department of Stomatology, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Zunyue Xu
- Department of Stomatology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Zheng Wang
- Department of Stomatology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Zhu
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China,CONTACT Qiang Zhu Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai200433, China
| |
Collapse
|
11
|
Bak J, Lee SJ, Kim TW, Hwang S, Park MJ, Arunachalam R, Yoo E, Park MH, Choi YS, Kim HK. Schisandrol A and gomisin N from Schisandra chinensis extract improve hypogonadism via anti-oxidative stress in TM3 Leydig cells. Nutr Res Pract 2023; 17:1-12. [PMID: 36777801 PMCID: PMC9884586 DOI: 10.4162/nrp.2023.17.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/27/2022] [Accepted: 06/22/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND/OBJECTIVES Male hypogonadism is a condition where the body does not produce enough testosterone and significantly impacts health. Age, obesity, genetics, and oxidative stress are some physiological factors that may contribute to testosterone deficiency. Previous studies have shown many pharmacological benefits of Schisandra chinensis (S. chinensis) Baillon as an anti-inflammatory and antioxidant. However, the molecular mechanism of attenuating hypogonadism is yet to be well established. This research was undertaken to study the effects of S. chinensis extract (SCE) on testosterone deficiency. MATERIALS/METHODS S. chinensis fruit was pulverized and extracted using 60% aqueous ethanol. HPLC analysis was performed to analyze and quantify the lignans of the SCE. RESULTS The 2,2-diphenyl-2-picrylhydrazyl and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) scavenging assays confirmed that the SCE and its major lignans (schisandrol A and gomisin N) inhibit oxidative stress. Effects of SCE analysis on the testosterone level under oxidative stress conditions revealed that both schisandrol A and gomisin N were able to recover the lowered testosterone levels. Through mRNA expression of TM3 Leydig cell, we observed that the SCE lignans were able to induce the enzymes involved in testosterone biosynthesis-related genes such as 3β-HSD4 (P < 0.01 for SCE, and P < 0.001 for schisandrol A and gomisin N), 17β-HSD3 (P < 0.001 for SCE, schisandrol A and gomisin N), and 17, 20-desmolase (P < 0.01 for schisandrol A, and P < 0.001 for SCE and gomisin N). CONCLUSIONS These results support that SCE and its active components could be potential therapeutic agents for regulating and increasing testosterone production.
Collapse
Affiliation(s)
- Jia Bak
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Seung Ju Lee
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Tae Won Kim
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Seonhwa Hwang
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Min Ju Park
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Rohith Arunachalam
- College of Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA
| | - Eunsoo Yoo
- College of Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA
| | - Min Hi Park
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Yun-Sik Choi
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| | - Hye Kyung Kim
- College of Pharmacy, Kyungsung University, Busan 48434, Korea
| |
Collapse
|
12
|
Zhou JH, Zhu XL, Li MY, Luo YW, Yang ZM, Wang L, Tong T, Qin SL, Liu BL, Chen BH, Hu W. Local adrenomedullin gene delivery inhibits Leydig cell dysfunction by rescuing steroidogenic enzymes in vivo. Andrologia 2022; 54:e14545. [PMID: 35942817 DOI: 10.1111/and.14545] [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: 03/18/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022] Open
Abstract
Adrenomedullin (ADM) has beneficial effects on Leydig cells under pathological conditions, including lipopolysaccharide (LPS)-induced orchitis. Our previous studies demonstrated that ADM exerts a restorative effect on steroidogenesis in LPS-treated primary rat Leydig cells by attenuating oxidative stress, inflammation and apoptosis. In this study, we aim to investigate whether ADM inhibits Leydig cell dysfunction by rescuing steroidogenic enzymes in vivo. Rats were administered with LPS and injected with Ad-ADM, an adeno-associated virus vector that expressed ADM. Then, rat testes were collected for 3β-hydroxysteroid dehydrogenase (3β-HSD) immunofluorescence staining. Steroidogenic enzymes or steroidogenic regulatory factors or protein, including steroidogenic factor-1 (SF-1), liver receptor homologue-1 (LRH1), Nur77, steroidogenic acute regulatory protein (StAR), cytochrome P450 cholesterol side chain cleavage enzyme (P450scc), 3β-HSD, cytochrome P450 17α-hydroxylase/17, 20 lyase (CYP17) and 17β-hydroxysteroid dehydrogenase (17β-HSD), were detected via gene expression profiling and western blot analysis. Plasma testosterone concentrations were measured. Results showed that ADM may inhibit Leydig cell dysfunction by rescuing steroidogenic enzymes and steroidogenic regulatory factors in vivo. The reduction in the number of Leydig cells after LPS exposure was reversed by ADM. ADM rescued the gene or protein levels of SF-1, LRH1, Nur77, StAR, P450scc, 3β-HSD, CYP17 and 17β-HSD and plasma testosterone concentrations. To summarize ADM could rescue some important steroidogenic enzymes, steroidogenic regulatory factors and testosterone production in Leydig cells in vivo.
Collapse
Affiliation(s)
- Jian-Hua Zhou
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xia-Lian Zhu
- Department of Nuclear Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ming-Yong Li
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - You-Wen Luo
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhi-Min Yang
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Wang
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Tao Tong
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Song-Lin Qin
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo-Long Liu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Bing-Hai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Hu
- Department of Andrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
13
|
Qian P, Wang Q, Wang FZ, Dai HB, Wang HY, Gao Q, Zhou H, Zhou YB. Adrenomedullin Improves Cardiac Remodeling and Function in Obese Rats with Hypertension. Pharmaceuticals (Basel) 2022; 15:ph15060719. [PMID: 35745637 PMCID: PMC9227996 DOI: 10.3390/ph15060719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to determine whether adrenomedullin (ADM, 7.2 μg/kg/day, ip), an important endogenous active peptide, has a protective role in cardiac remodeling and function in obesity-related hypertension (OH) rats. A high-fat diet (HFD) was used to induce OH for 20 weeks. H9c2 cells incubated with palmitate (PA, 200 μM) to mimic high free fatty acid in obesity were used as an in vitro model. In OH rats, ADM not only decreased body weight (BW) and blood pressure (BP) but also improved systemic inflammation and oxidative stress. Moreover, ADM still had a greater inhibitory effect on local inflammation and oxidative stress in the hearts of OH rats, and the same anti-inflammatory and antioxidant effects were also confirmed in PA-treated H9c2 cells. The ADM receptor antagonist or Akt inhibitor effectively attenuated the inhibitory effects of ADM on inflammation and oxidative stress in PA-stimulated H9c2 cells. Furthermore, ADM application effectively normalized heart function, and hematoxylin-eosin and Masson staining and collagen volume fraction results showed that ADM improved cardiac remodeling in hearts of OH rats. ADM attenuated cardiac inflammation and oxidative stress via the receptor-Akt pathway, which involves the improvement of cardiac remodeling and function in OH rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ye-Bo Zhou
- Correspondence: ; Tel./Fax: +86-25-8686-9351
| |
Collapse
|
14
|
Alehagen U, Johansson P, Svensson E, Aaseth J, Alexander J. Improved cardiovascular health by supplementation with selenium and coenzyme Q10: applying structural equation modelling (SEM) to clinical outcomes and biomarkers to explore underlying mechanisms in a prospective randomized double-blind placebo-controlled intervention project in Sweden. Eur J Nutr 2022; 61:3135-3148. [PMID: 35381849 PMCID: PMC9363287 DOI: 10.1007/s00394-022-02876-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
Purpose Selenium and coenzyme Q10 have synergistic antioxidant functions. In a four-year supplemental trial in elderly Swedes with a low selenium status, we found improved cardiac function, less cardiac wall tension and reduced cardiovascular mortality up to 12 years of follow-up. Here we briefly review the main results, including those from studies on biomarkers related to cardiovascular risk that were subsequently conducted. In an effort, to explain underlying mechanisms, we conducted a structured analysis of the inter-relationship between biomarkers. Methods Selenium yeast (200 µg/day) and coenzyme Q10 (200 mg/ day), or placebo was given to 443 elderly community-living persons, for 48 months. Structural Equation Modelling (SEM) was used to investigate the statistical inter-relationships between biomarkers related to inflammation, oxidative stress, insulin-like growth factor 1, expression of microRNA, fibrosis, and endothelial dysfunction and their impact on the clinical effects. The main study was registered at Clinicaltrials.gov at 30th of September 2011, and has the identifier NCT01443780. Results In addition to positive clinical effects, the intervention with selenium and coenzyme Q10 was also associated with favourable effects on biomarkers of cardiovascular risk. Using these results in the SEM model, we showed that the weights of the first-order factors inflammation and oxidative stress were high, together forming a second-order factor inflammation/oxidative stress influencing the factors, fibrosis (β = 0.74; p < 0.001) and myocardium (β = 0.65; p < 0.001). According to the model, the intervention impacted fibrosis and myocardium through these factors, resulting in improved cardiac function and reduced CV mortality. Conclusion Selenium reduced inflammation and oxidative stress. According to the SEM analysis, these effects reduced fibrosis and improved myocardial function pointing to the importance of supplementation in those low on selenium and coenzyme Q10.
Collapse
Affiliation(s)
- Urban Alehagen
- Division of Cardiovascular Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, 581 85, Linköping, Sweden.
| | - Peter Johansson
- Department of Health, Medicine and Caring Sciences, Linköping University, 601 74, Norrköping, Sweden
| | | | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, 2381, Brumunddal, Norway.,Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, 2418, Elverum, Norway
| | - Jan Alexander
- Norwegian Institute of Public Health, 0403, Oslo, Norway
| |
Collapse
|
15
|
Sağraç D, Şenkal S, Hayal TB, Demirci S, Şişli HB, Asutay AB, Doğan A. Protective role of Cytoglobin and Neuroglobin against the Lipopolysaccharide (LPS)-induced inflammation in Leydig cells ex vivo. Reprod Biol 2022; 22:100595. [PMID: 35121559 DOI: 10.1016/j.repbio.2021.100595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 11/21/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022]
Abstract
Leydig cells are responsible for testosterone production in male testis upon stimulation by luteinizing hormone. Inflammation and oxidative stress related Leydig cell dysfunction is one of the major causes of male infertility. Cytoglobin (CYGB) and Neuroglobin (NGB) are two globin family member proteins which protect cells against oxidative stress. In the current study, we established a Lipopolysaccharide (LPS)-induced inflammation model in TM3 Leydig cell culture to study the function of CYGB and NGB proteins under inflammatory conditions. CYGB and NGB were downregulated using siRNA and shRNA based experimental strategies. Overexpression was conducted using lentiviral pLenti-III-CYGB-2A-GFP, and pLenti-III-NGB-2A-GFP vector systems. As testicular macrophages regulate immune function upon inflammation and steroidogenesis of Leydig cells, we generated direct/indirect co-culture systems of TM3 and mouse macrophage (RAW264.7) cells ex vivo. Downregulation of CYGB and NGB induced nitride oxide (NO) release, blocked cell cycle progression, reduced testosterone production and increased inflammatory and apoptotic pathway gene expression in the presence and absence of LPS. On the other hand, CYGB and NGB overexpression reduced TNFα and COX-2 protein expressions and increased the expression of testosterone biogenesis pathway genes upon LPS stimulation. In addition, CYGB and NGB overexpression upregulated testosterone production. The present study successfully established an inflammatory interaction model of TM3 and RAW264.7 cells. Suppression of CYGB and NGB in TM3 cells changed macrophage morphology, enhanced macrophage cell number and NO release in co-culture experiments upon LPS exposure. In summary, these results demonstrate that globin family members might control LPS induced inflammation by regulating apoptotic mechanisms and macrophage response.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayla Burçin Asutay
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
16
|
Liu C, Hao K, Liu Z, Liu Z, Guo N. Epigallocatechin gallate (EGCG) attenuates staphylococcal alpha-hemolysin (Hla)-induced NLRP3 inflammasome activation via ROS-MAPK pathways and EGCG-Hla interactions. Int Immunopharmacol 2021; 100:108170. [PMID: 34562843 DOI: 10.1016/j.intimp.2021.108170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022]
Abstract
Alpha-hemolysin (Hla), the virulence factor secreted by Staphylococcus aureus (S. aureus), plays a critical role in infection and inflammation, which is a severe health burden worldwide. Therefore, it is necessary to develop a drug against Hla. Epigallocatechin gallate (EGCG), a polyphenol extracted from green tea, has excellent anti-inflammatory activity. In this study, we investigated the inhibitory effect of EGCG on Hla-induced NLRP3 inflammasome activation in vitro and in vivo and elucidated the potential molecular mechanism. We found that EGCG attenuated the hemolysis of Hla by inhibiting its secretion. Besides, EGCG significantly decreased overproduction of ROS and activation of MAPK signaling pathway induced by Hla, thereby markedly attenuating the expression of NLRP3 inflammasome-related proteins in THP-1 cells. Notably, EGCG could spontaneously bind to Hla with affinity constant of 1.71 × 10-4 M, thus blocking the formation of the Hla heptamer. Moreover, Hla-induced expression of NLRP3, ASC and caspase-1 protein and generation of IL-1β and IL-18 in the damaged liver tissue of mice were also significantly suppressed by EGCG in a dose-dependent manner. Collectively, EGCG could be a promising candidate for alleviating Hla-induced the activation of NLRP3 inflammasome, depending on ROS mediated MAPK signaling pathway, and inhibition of Hla secretion and heptamer formation. These findings will enlighten the applications of EGCG to reduce the S. aureus infection by targeting Hla in food and related pharmaceutical fields.
Collapse
Affiliation(s)
- Chunmei Liu
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Kun Hao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Zuojia Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zonghui Liu
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Na Guo
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
17
|
Luo P, Feng X, Deng R, Wang F, Zhang Y, Li X, Zhang M, Wan Z, Xiang AP, Xia K, Gao Y, Deng C. An autofluorescence-based isolation of Leydig cells for testosterone deficiency treatment. Mol Cell Endocrinol 2021; 535:111389. [PMID: 34229003 DOI: 10.1016/j.mce.2021.111389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 11/20/2022]
Abstract
Effective procedures for the purification of Leydig cells (LCs) can facilitate functional studies and transplantation therapies. However, current methods to purify LCs from testes are still far from satisfactory. Here, we found that testicular autofluorescence existed in the interstitium along with the gradual maturation of LCs from birth to adulthood. These autofluorescent cells were further isolated by fluorescence-activated cell sorting (FACS) and determined to be composed of LCs and macrophages. To further purify LCs, we combined two fluorescence channels of FACS and successfully separated LCs and macrophages. Of note, we confirmed that the obtained LCs not only possessed high purity, viability and quantity but also had intact steroidogenic activity and excellent responsiveness to luteinizing hormone. Moreover, subcutaneous transplantation of isolated LCs could alleviate the symptoms of testosterone deficiency in castrated mice. In summary, we established an effective autofluorescence-based method for isolating LCs. This method will aid in the future success of using LCs for basic and translational applications.
Collapse
Affiliation(s)
- Peng Luo
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Xin Feng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ronghai Deng
- Department of Organ Transplantation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fulin Wang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yadong Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangping Li
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zi Wan
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kai Xia
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
18
|
Cao J, Lu M, Yan W, Li L, Ma H. Dehydroepiandrosterone alleviates intestinal inflammatory damage via GPR30-mediated Nrf2 activation and NLRP3 inflammasome inhibition in colitis mice. Free Radic Biol Med 2021; 172:386-402. [PMID: 34182071 DOI: 10.1016/j.freeradbiomed.2021.06.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/27/2023]
Abstract
Dehydroepiandrosterone (DHEA) is a popular dietary supplement that has anti-inflammatory, anti-oxidant and immune-regulating role; meanwhile, it also can effective in the protection of inflammation diseases such as inflammatory bowel disease (IBD), but the underlying mechanisms remain elusive. Here, we demonstrated that DHEA inhibits excessive inflammation response and enhances gut barrier function via activating the G protein-coupled receptor 30 (GPR30). GPR30-induced the ERK phosphorylation and p62 accumulation led to the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, which subsequently inhibited the reactive oxygen species (ROS) overproduction and finally alleviated the intestinal barrier dysfunction. Furthermore, DHEA blocked the p38-induced NLRP3 inflammasome activation in both LPS-stimulated colon epithelial cells and macrophages. In addition, in vivo results showed that DHEA and GPR30 agonist G1 attenuated inflammatory responses and gut barrier dysfunction in colitis mice, while the GPR30 specific inhibitor G15 abrogated these beneficial effects of DHEA. Cumulatively, our study unveiled that DHEA is an effective anti-inflammatory agent and suggested that GPR30 could as a potential target for the treatment of IBD.
Collapse
Affiliation(s)
- Ji Cao
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Miaomiao Lu
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Weiyuan Yan
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Longlong Li
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
19
|
Sun J, Gao L, Huang S, Wang L, Yang W, Zhang T, Jin Y, Song L. CLec-TM1-ERK-GSK3β Pathway Regulates Vibrio splendidus-Induced IL-17 Production in Oyster. THE JOURNAL OF IMMUNOLOGY 2021; 207:640-650. [PMID: 34193596 DOI: 10.4049/jimmunol.2100007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/16/2021] [Indexed: 11/19/2022]
Abstract
C-type lectins are a family of pattern recognition receptors that recognize microbial components and subsequently activate the signaling cascade to induce the production of proinflammatory cytokines. In the current study, the homologs of ERK (named as CgERK) and GSK3β (named as CgGSK3β) and a novel C-type lectin with a transmembrane domain (named as CgCLec-TM1) were identified from oyster Crassostrea gigas CgCLec-TM1 was able to bind Escherichia coli and Vibrio splendidus through its carbohydrate recognition domain and then activated CgERK by inducing its phosphorylation. The activated CgERK interacted with CgGSK3β to phosphorylate it at Ser9, which eventually induced the expressions of CgIL-17-1 and CgIL-17-5. The interaction between CgERK and CgGSK3β, as well as the phosphorylation of CgGSK3β, could be inhibited by ERK inhibitor (PD98059) to reduce the expressions of CgIL-17-1 and CgIL-17-5. CgGSK3β in oyster was proposed as a new substrate of CgERK. The results defined a CLec-TM1-ERK-GSK3β signaling pathway in oyster, which was activated by V. splendidus and then induced CgIL-17 productions.
Collapse
Affiliation(s)
- Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Lei Gao
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Shu Huang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Southern Laboratory of Ocean Science and Engineering, Zhuhai, China; and.,Dalian Key Laboratory of Aquatic Animal Disease Control, Dalian Ocean University, Dalian, China
| | - Wenwen Yang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Tong Zhang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Yingnan Jin
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China; .,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China.,Southern Laboratory of Ocean Science and Engineering, Zhuhai, China; and
| |
Collapse
|
20
|
Tanaka M, Kakihara S, Hirabayashi K, Imai A, Toriyama Y, Iesato Y, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Tanaka M, Cui N, Wei Y, Zhao Y, Aruga K, Yamauchi A, Murata T, Shindo T. Adrenomedullin-Receptor Activity-Modifying Protein 2 System Ameliorates Subretinal Fibrosis by Suppressing Epithelial-Mesenchymal Transition in Age-Related Macular Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:652-668. [PMID: 33385343 DOI: 10.1016/j.ajpath.2020.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 01/06/2023]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment. Anti-vascular endothelial growth factor drugs used to treat AMD carry the risk of inducing subretinal fibrosis. We investigated the use of adrenomedullin (AM), a vasoactive peptide, and its receptor activity-modifying protein 2, RAMP2, which regulate vascular homeostasis and suppress fibrosis. The therapeutic potential of the AM-RAMP2 system was evaluated after laser-induced choroidal neovascularization (LI-CNV), a mouse model of AMD. Neovascular formation, subretinal fibrosis, and macrophage invasion were all enhanced in both AM and RAMP2 knockout mice compared with those in wild-type mice. These pathologic changes were suppressed by intravitreal injection of AM. Comprehensive gene expression analysis of the choroid after LI-CNV with or without AM administration revealed that fibrosis-related molecules, including Tgfb, Cxcr4, Ccn2, and Thbs1, were all down-regulated by AM. In retinal pigment epithelial cells, co-administration of transforming growth factor-β and tumor necrosis factor-α induced epithelial-mesenchymal transition, which was also prevented by AM. Finally, transforming growth factor-β and C-X-C chemokine receptor type 4 (CXCR4) inhibitors eliminated the difference in subretinal fibrosis between RAMP2 knockout and wild-type mice. These findings suggest the AM-RAMP2 system suppresses subretinal fibrosis in LI-CNV by suppressing epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Masaaki Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Shinji Kakihara
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | | | - Akira Imai
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Yuichi Toriyama
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Yasuhiro Iesato
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Nanqi Cui
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yangxuan Wei
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yunlu Zhao
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kohsuke Aruga
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akihiro Yamauchi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toshinori Murata
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan.
| |
Collapse
|
21
|
El-Saka MH, Barhoma RA, Ibrahim RR, Elsaadany A, Alghazaly GM, Elshwaikh S, Marea KE, Madi NM. Potential effect of adrenomedullin on metabolic and endocrinal dysfunctions in the experimentally induced polycystic ovary: Targeting implication of endoplasmic reticulum stress. J Biochem Mol Toxicol 2021; 35:e22725. [PMID: 33491863 DOI: 10.1002/jbt.22725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/26/2020] [Accepted: 01/09/2021] [Indexed: 02/06/2023]
Abstract
This study investigated the potential effect of adrenomedullin (ADM) on metabolic and endocrinal dysfunctions in experimentally induced polycystic ovary. Twenty-four female Wistar rats were allocated into three groups: control; polycystic ovary syndrome (PCOS) in which PCOS was induced by letrozole, orally in a dose of 1 mg/kg once daily for 3 weeks; and ADM group in which ADM was injected intraperitonally in a dose of 3.5/μg/twice daily for 4 weeks. At the end of the experimental period, the serum sex hormone profile, ADM, fasting glucose, insulin, homeostatic model assessment of insulin resistance, and lipid parameters were determined. Ovarian tissue homogenates were used to determine malondialdehyde, total antioxidant capacity, glutathione peroxidase activity, tumor necrosis factor α, interleukin 6, B cell lymphoma-2 (Bcl-2), and Bcl-2 associated X protein. The profibrotic growth factors, including transforming growth factor β1 and connective tissue growth factor, were determined; and also, the relative gene expression of endoplasmic reticulum (ER) stress, including (Xbox-binding protein-1 [XBP-1], activating transcription factor 6 [ATF6], and homologous protein [CHOP]), serine/threonine kinase 1 (Akt1), phosphatidylinositol 3-kinase (PI3K), and peroxisome proliferator-activated receptor γ (PPAR-γ) were determined. Finally, histopathological analysis of the ovaries was evaluated. PCOS group exhibited increased ER stress, suppressing of PI3K/Akt1 and PPAR-γ pathways, imbalance of sex hormonal profile, hyperglycemia, insulin resistance, dyslipidemia, increased profibrotic factors, and abnormal ovarian histopathological picture, while ADM treatment alleviated these disturbances occurring in the PCOS model. We concluded that ADM mitigated PCOS via attenuating the ER stress, in addition to activation of PI3K/Akt1 and PPAR-γ pathways, its antioxidant, anti-inflammatory, antiapoptotic, and antifibrotic properties.
Collapse
Affiliation(s)
- Mervat H El-Saka
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ramez A Barhoma
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rowida R Ibrahim
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira Elsaadany
- Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ghada M Alghazaly
- Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shereef Elshwaikh
- Gynecology and Obstetrics, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Karima E Marea
- Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nermin M Madi
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
22
|
Dai H, Wang F, Kang Y, Sun J, Zhou H, Gao Q, Li Z, Qian P, Zhu G, Zhou Y. Adrenomedullin Attenuates Inflammation in White Adipose Tissue of Obese Rats Through Receptor-Mediated PKA Pathway. Obesity (Silver Spring) 2021; 29:86-97. [PMID: 32985779 PMCID: PMC7821304 DOI: 10.1002/oby.23012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Adrenomedullin (ADM) possesses therapeutic potential for inflammatory diseases. Consequently, the effects of ADM on inflammation in visceral white adipose tissue (vWAT) of obese rats or in adipocytes were explored in this study. METHODS Male rats were fed a high-fat diet for 12 weeks to induce obesity, and obese rats were implanted with osmotic minipumps providing constant infusion of ADM (300 ng/kg per hour) and continued to be fed a high-fat diet for 4 weeks. RESULTS When compared with the control group, endogenous protein expression of ADM and ADM receptors in vWAT and in lipopolysaccharide (LPS)-treated adipocytes was markedly increased. ADM significantly decreased the protein expression of the inflammatory mediators TNFα, IL-1β, cyclooxygenase-2, and inducible nitric oxide synthase in vWAT of obese rats and in adipocytes stimulated by LPS. It also inhibited the activation of the inflammatory signaling pathways MAPK and NF-κB induced by LPS in adipocytes. These effects of ADM in adipocytes were inhibited by the administration of ADM receptor antagonist and cAMP-dependent protein kinase (PKA) activation inhibitor. CONCLUSIONS ADM can inhibit inflammation in WAT in obesity, which may be mediated by the activation of ADM receptors and PKA.
Collapse
Affiliation(s)
- Hang‐Bing Dai
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Fang‐Zheng Wang
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Ying Kang
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Jing Sun
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Hong Zhou
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Qing Gao
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Zhen‐Zhen Li
- Department of CardiologyBenQ Medical CenterThe Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Pei Qian
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Guo‐Qing Zhu
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Ye‐Bo Zhou
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| |
Collapse
|
23
|
MIF inhibitor, ISO-1, attenuates human pancreatic cancer cell proliferation, migration and invasion in vitro, and suppresses xenograft tumour growth in vivo. Sci Rep 2020; 10:6741. [PMID: 32317702 PMCID: PMC7174354 DOI: 10.1038/s41598-020-63778-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
This study sought to investigate the biological effects of specific MIF inhibitor, ISO-1, on the proliferation, migration and invasion of PANC-1 human pancreatic cells in vitro, and on tumour growth in a xenograft tumour model in vivo. The effect of ISO-1 on PANC-1 cell proliferation was examined using CCK-8 cell proliferation assay. The effect of ISO-1 on collective cell migration and recolonization of PANC-1 cells was evaluated using the cell-wound closure migration assay. The effect of ISO-1 on the migration and invasion of individual PANC-1 cells in a 3-dimensional environment in response to a chemo-attractant was investigated through the use of Transwell migration/invasion assays. Quantitative real time PCR and western blot analyses were employed to investigate the effects of ISO-1 on MIF, NF-κB p65 and TNF-α mRNA and protein expression respectively. Finally, a xenograft tumor model in BALB/c nude mice were used to assess the in vivo effects of ISO-1 on PANC-1-induced tumor growth. We found high expression of MIF in pancreatic cancer tissues. We demonstrated that ISO-1 exerts anti-cancer effects on PANC-1 cell proliferation, migration and invasion in vitro, and inhibited PANC-1 cell-induced tumour growth in xenograft mice in vivo. Our data suggests that ISO-1 and its derivative may have potential therapeutic applications in pancreatic cancer.
Collapse
|
24
|
Bahar L, Sucu N, Eras N, Bagdatoglu OT, Yildirim M. Adrenomedullin expression in aortic artery wall of diabetic rats given alpha lipoic acid. Pharmacol Rep 2020; 72:912-921. [PMID: 32103464 DOI: 10.1007/s43440-020-00082-6] [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: 05/03/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Diabetes mellitus (DM) is a major health problem predisposing to cardiovascular diseases. The aim of this study was to investigate the effects of alpha lipoic acid (ALA) on both the arterial wall of diabetic rats and the adrenomedullin (ADM) gene expression. METHODS Twenty-four Wistar Albino rats were divided into three groups as Control, DM + S, and DM + ALA. For DM model, a single dose of 40 mg/kg streptozotocin, for DM + ALA group, 100 mg/kg/day/4 weeks was administered. Hematoxylin & Eosin (H&E) staining was done and vascular endothelial growth factor (VEGF) was detected by immunohistochemical analysis in the artery wall. Total damage score of vessel wall (endothelial cell damage, media layer smooth muscle cell damage, and internal elastic lamina damage) and H score (immunoreactivity intensity) were calculated. Expression of ADM gene was measured by qRT-PCR. RESULTS In DM + S group, Total damage score of vessel wall were detected by light microscopy. There were statistically significant differences between the groups Control/DM + S and DM + S/DM + ALA in terms of the vessel total damage score and H score (p < 0.005). ADM expression was increased threefold in both DM + S and DM + ALA groups compared to the control group (p < 0.05). CONCLUSIONS ALA may have positive effect on the vessel damage in diabetic rats. However, no significant decrease in ADM expression levels was observed in diabetic rats after ALA administration and we considered that the protective effect of ALA is independent of adrenomedullin. Further studies with different doses and durations of ALA administrations are required to investigate the changes in ADM expression.
Collapse
Affiliation(s)
- Leyla Bahar
- Vocational School of Health Services, Mersin University, Ciftlikkoy Campuse, 33343 Yenisehir, Mersin, Turkey.
| | - Nehir Sucu
- Department of Cardiovascular Surgery, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Nazan Eras
- Department of Medical Genetics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ozlen Tubay Bagdatoglu
- Vocational School of Health Services, Mersin University, Ciftlikkoy Campuse, 33343 Yenisehir, Mersin, Turkey
| | - Metin Yildirim
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| |
Collapse
|
25
|
Sun Y, Xiong X, Wang X. HIF1α/miR-199a/ADM feedback loop modulates the proliferation of human dermal microvascular endothelial cells (HDMECs) under hypoxic condition. Cell Cycle 2019; 18:2998-3009. [PMID: 31537150 DOI: 10.1080/15384101.2019.1666611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hypoxia-inducible factor 1α (HIF1α) plays a protective role in the hypoxia-induced cellular injury. In the present study, we attempted to investigate the role and mechanism of HIF1αin human dermal microvascular endothelial cells (hDMECs), a common-used cell model for researches on the hypoxia-induced injury during skin wounds healing. As revealed by ChIP and online tools prediction and confirmed by luciferase reporter and ChIP assays, HIF1A can bind to the promoter regions of ADM and miR-199a, while miR-199a directly binds to the 3'UTR of HIF1A and ADM. Hypoxia stress induces HIF1α and ADM expression while inhibits miR-199a expression. Under hypoxic condition, HIF1α knockdown increases the nucleus translocation of p65 and the release of TNF-α and IL-8, inhibits the proliferation and migration, while promotes the cellular permeability in HDMECs upon hypoxic stress, while ADM overexpression and miR-199a inhibition exerted an opposite effect on HDMECs. ADM overexpression or miR-199a inhibition could partially reverse the effect of HIF1A knockdown under hypoxia. In summary, we demonstrate a feedback loop consists of HIF1α, miR-199a, and ADM which protect HDMECs from hypoxia-induced cellular injury by modulating the inflammation response, cell proliferation, migration and permeability in HDMECs.
Collapse
Affiliation(s)
- Yang Sun
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| | - Xiang Xiong
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| | - Xiancheng Wang
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| |
Collapse
|
26
|
Zheng Y, Hu G, Wu W, Zhao Z, Meng S, Fan L, Song C, Qiu L, Chen J. Transcriptome analysis of juvenile genetically improved farmed tilapia (Oreochromis niloticus) livers by dietary resveratrol supplementation. Comp Biochem Physiol C Toxicol Pharmacol 2019; 223:1-8. [PMID: 31028934 DOI: 10.1016/j.cbpc.2019.04.011] [Citation(s) in RCA: 6] [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: 03/14/2019] [Revised: 04/18/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Abstract
Here we used RNA-Seq to explore the transcriptomic response and specific involvement of hepatic mRNA of juvenile Oreochromis niloticus (GIFT) as a result of dietary resveratrol supplementation (0.05 g/kg RES). More than 24,513,018 clean reads were reference genome guided assembly into 23,417 unigenes. 12,596 unigenes (29.64%) were annotated to GO database. There were 5, 179 and 1526 genes significantly differentially expressed genes at 15, 30 and 45 d respectively, and 8 KEGG pathways were enriched associated with this immune response. Hyperemia and compressed hepatic sinusoid, fibrosis of liver cell and abnormal hepatic epidermal cell revealed by H&E and SEM analysis respectively. Genes related with cytokine production (il12rb2, scfr), immune system (ig8l, hlfl, cd226, prf1l), autophagy regulation (atg4b), foxo signaling (ccnb2), steroid hormone biosynthesis (cyp3a40), fatty acid metabolism (scd1), metabolism (cacna1b) have been significantly decreased, while genes associated with such pathways above (leap-2, prdx4, mb, homer1, mif, sat1, cytbc1_8) and the pathway of protein processing in endoplasmic reticulum (cne1, tram1) have been significantly increased. These findings suggested RES activated some immune and biological process-related genes to enhance GIFT's innate immunity. It also suggested high concentration addition or long-time administration may bring negative effect in tilapia liver.
Collapse
Affiliation(s)
- Yao Zheng
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Gengdong Hu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Wei Wu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Zhixiang Zhao
- Wuxi Fishery College, Nanjing Agricultural University, Wuxi, Jiangsu 214081, PR China
| | - Shunlong Meng
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Limin Fan
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Chao Song
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Liping Qiu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China
| | - Jiazhang Chen
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Evironment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Environmental Factors(Wuxi), Ministry of Agriculture, Wuxi, Jiangsu 214081, PR China; Wuxi Fishery College, Nanjing Agricultural University, Wuxi, Jiangsu 214081, PR China; Key Laboratory of Control of Quality and Safety for Aquatic Products, Ministry of Agriculture, PR China.
| |
Collapse
|
27
|
Li MY, Zhu XL, Zhao BX, Shi L, Wang W, Hu W, Qin SL, Chen BH, Zhou PH, Qiu B, Gao Y, Liu BL. Adrenomedullin alleviates the pyroptosis of Leydig cells by promoting autophagy via the ROS-AMPK-mTOR axis. Cell Death Dis 2019; 10:489. [PMID: 31222000 PMCID: PMC6586845 DOI: 10.1038/s41419-019-1728-5] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 01/03/2023]
Abstract
Adrenomedullin (ADM) exerts anti-oxidant, anti-inflammatory and anti-apoptotic effects in Leydig cells. However, the role and mechanism of ADM in the pyroptosis of Leydig cells are poorly understood. This study first showed the protective effects of ADM on the pyroptosis and biological functions of Leydig cells exposed to lipopolysaccharide (LPS) by promoting autophagy. Primary rat Leydig cells were treated with various concentrations of LPS and ADM, together with or without N-acetyl-L-cysteine (NAC) or 3-methyladenine (3-MA). Cell proliferation was detected through CCK-8 and BrdU incorporation assays, and ROS level was measured with the DCFDA assay. Real-time PCR, western blot, immunofluorescence, transmission electron microscopy, TUNEL and flow cytometry were performed to examine ADM's effect on the pyroptosis, autophagy and steroidogenic enzymes of Leydig cells and AMPK/mTOR signalling. Like NAC, ADM dose-dependently reduced LPS-induced cytotoxicity and ROS overproduction. ADM also dose-dependently ameliorated LPS-induced pyroptosis by reversing the increased expression of NLRP3, ASC, caspase-1, IL-1β, IL-18, GSDMD, caspase-3, caspase-7, TUNEL-positive and PI and active caspase-1 double-stained positive rate, DNA fragmentation and LDH concentration, which could be rescued via co-incubation with 3-MA. ADM dose-dependently increased autophagy in LPS-induced Leydig cells, as confirmed by the increased expression of LC3-I/II, Beclin-1 and ATG-5; decreased expression of p62 and autophagosomes formation; and increased LC3-II/LC3-I ratio. However, co-treatment with 3-MA evidently decreased autophagy. Furthermore, ADM dose-dependently rescued the expression of steroidogenic enzymes, including StAR, P450scc, 3β-HSD and CYP17, and testosterone production in LPS-induced Leydig cells. Like rapamycin, ADM dose-dependently enhanced AMPK phosphorylation but reduced mTOR phosphorylation in LPS-induced Leydig cells, which could be rescued via co-incubation with 3-MA. In addition, pyroptosis was further decreased, and autophagy was further promoted in LPS-induced Leydig cells upon co-treatment with ADM and rapamycin. ADM may protect the steroidogenic functions of Leydig cells against pyroptosis by activating autophagy via the ROS-AMPK-mTOR axis.
Collapse
Grants
- Hunan Natural Science Foundation, Hunan, China (Grant No.: 2019JJ40269), Health and Family Planning Research Project of Hunan Province, Changsha, China (Grant No.: B2017051)
- National Science Foundation of China, Beijing, China (Grant No.: 81401190)
- Social Development Foundation of Zhenjiang, Zhenjiang, China (Grant No.: SH2016031)
- National Science Foundation of China, Beijing, China (Grant No.: 81501921),Science and Technology Project of Wuhan, China (Grant No.: 2016060101010045)
- National Science Foundation of China, Beijing, China (Grant No.: 81602241)
- National Science Foundation of China, Beijing, China (Grant Nos.: 81471449,81871110 and 81671449),Guangdong Province Natural Science Foundation, Guangzhou, China (Grant No.: 2015A030313141), Guangdong Province Science and Technology Project, Guangzhou, China (Grant Nos.: 2016B030230001 and 2016A040403113), Key Scientific and Technological Program of Guangzhou City, Guangzhou, China (Grant No.: 201604020189)
Collapse
Affiliation(s)
- Ming-Yong Li
- Department of Urology, The First Affiliated Hospital of University of South China, No. 69 Chuan Shan Road, Hengyang, 421001, Hunan Province, China
| | - Xia-Lian Zhu
- Department of Hand Surgery, Affiliated Nanhua Hospital of University of South China, No. 336 Dong Feng South Road, Hengyang, 421002, Hunan Province, China
| | - Bi-Xia Zhao
- Department of Urology, Affiliated Nanhua Hospital of University of South China, No. 336 Dong Feng South Road, Hengyang, 421002, Hunan Province, China
| | - Lei Shi
- Department of Oncology, Renmin Hospital of Wuhan University, No. 238 Liberation Road, Wuhan, 430060, Hubei Province, China
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Wei Hu
- Department of Andrology, The First Affiliated Hospital of University of South China, No. 69 Chuan Shan Road, Hengyang, 421001, Hunan Province, China.
| | - Song-Lin Qin
- Department of Andrology, The First Affiliated Hospital of University of South China, No. 69 Chuan Shan Road, Hengyang, 421001, Hunan Province, China.
| | - Bing-Hai Chen
- Department of Urology, Affiliated Hospital of Jiangsu University, No. 438 Liberation Road, Zhenjiang, 212000, Jiangsu Province, China.
| | - Pang-Hu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, No. 238 Liberation Road, Wuhan, 430060, Hubei Province, China
| | - Bo Qiu
- Department of Orthopedics, Renmin Hospital of Wuhan University, No. 238 Liberation Road, Wuhan, 430060, Hubei Province, China
| | - Yong Gao
- Reproductive Medicine Centre, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Second Zhongshan Road, Guangzhou, 510080, Guangdong Province, China
| | - Bo-Long Liu
- Department of Andrology, The First Affiliated Hospital of University of South China, No. 69 Chuan Shan Road, Hengyang, 421001, Hunan Province, China
| |
Collapse
|
28
|
Dopamine Alters Lipopolysaccharide-Induced Nitric Oxide Production in Microglial Cells via Activation of D1-Like Receptors. Neurochem Res 2019; 44:947-958. [PMID: 30659504 DOI: 10.1007/s11064-019-02730-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023]
Abstract
Dopamine (DA) is important in the maintenance of normal nervous system function. DA is the target of multiple drugs, and it induces critical alterations in immune cells. However, these impacts are controversial, and the mechanism remains unclear. In the present study, we treated BV-2 microglial cells and primary microglia with DA and measured the changes in cytokines. We also identified the expression of DA receptors (DRs) using confocal and immunofluorescent microscopy. Specific agonists and antagonists of D1-like DRs (D1DR and D5DR) were used to observe alterations in cytokines. Western blot and siRNA interference were performed to investigate the involvement of the downstream signaling molecules of DRs. We also measured changes in mitogen-activated protein kinases (MAPKs) and the nuclear factor-kappa B (NF-κB) signaling pathway and assessed their involvement using inhibitors. We found that DA alone produced no effects on IL-6, TNF-α or nitric oxide (NO) production, and it inhibited lipopolysaccharide (LPS)-induced NO in microglial cells. Microglia expressed a high abundance of D1-like DRs (D1DR and D5DR). The agonists inhibited NO production, and antagonists reversed the DA-induced suppression of NO. Adenylatec cyclase (AC), cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) mediated DA function, and cAMP-response element binding protein (CREB) was not involved. ERK1/2 and NF-κB, but not p-38 or JNK, played roles in DA-suppressed NO generation via altering inducible nitric oxide synthase (iNOS) transcription. These data illustrate that DA modulates LPS-induced NO production via the AC/cAMP-PKA-ERK1/2-NF-κB-iNOS axis in mouse microglia, and D1-like DRs are involved. The present study provides functional evidence for an essential role of DA in immunoregulation.
Collapse
|
29
|
Theas MS. Germ cell apoptosis and survival in testicular inflammation. Andrologia 2018; 50:e13083. [DOI: 10.1111/and.13083] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/13/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- María Susana Theas
- Cátedra II de Histología, Departamento de Biología Celular, Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED); CONICET-Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
30
|
Kuchakulla M, Masterson T, Arora H, Kulandavelu S, Ramasamy R. Effect of nitroso-redox imbalance on male reproduction. Transl Androl Urol 2018; 7:968-977. [PMID: 30505735 PMCID: PMC6256041 DOI: 10.21037/tau.2018.08.14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are byproducts of normal metabolic processes. They are necessary for normal cellular function and are kept in balance by antioxidant mechanisms. Alterations in levels of ROS and RNS can lead to nitroso-redox imbalance that in turn can negatively affect male reproduction. Strategies to decrease ROS/RNS involve evasion of exposures (smoking, meat intake, pollution, calorie-dense diet), managing lifestyle, and increasing the consumption of antioxidants (vitamin C, vitamin E, alpha-lipoic acid, taurine, quercetin). Targeted therapies focusing on nitroso-redox imbalance can be critical for treatment of male reproductive dysfunction. This review outlines endogenous and exogenous sources of ROS/RNS, adverse effect on male reproduction, and strategies to control nitroso-redox imbalance.
Collapse
Affiliation(s)
- Manish Kuchakulla
- 1Department of Urology, 2The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Thomas Masterson
- 1Department of Urology, 2The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Himanshu Arora
- 1Department of Urology, 2The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- 1Department of Urology, 2The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ranjith Ramasamy
- 1Department of Urology, 2The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
31
|
Lin Y, Zhang J, Cai J, Liang R, Chen G, Qin G, Han X, Yuan C, Liu Z, Li Y, Zou D, Mao Y. Systematic Analysis of Gene Expression Alteration and Co-Expression Network of Eukaryotic Initiation Factor 4A-3 in Cancer. J Cancer 2018; 9:4568-4577. [PMID: 30588240 PMCID: PMC6299400 DOI: 10.7150/jca.27655] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose: Eukaryotic initiation factor 4A-3 (EIF4A3) is an RNA-binding protein (RBP) that is a core component of the exon junction complex (EJC). It has been identified as an important player in post-transcriptional regulation processes. Recently, investigations have focused on EIF4A3 dysfunction in carcinogenesis. The present study aims to determine whether EIF4A3 can serve as a prognostic marker and potential regulatory mechanism in human cancers. Materials and methods: EIF4A3 expression in various cancers was assessed using Oncomine. The Correlation between EIF4A3 expression and patient survival was evaluated using PrognoScan. EIF4A3 mutations in various cancers were investigated using cBioPortal. EIF4A3 co-expression networks in various cancers were established using Coexpedia. Finally, we analyzed potential functional roles of EIF4A3 using Gene Ontology and pathway enrichment analyses by FunRich V3. Results: EIF4A3 was overexpressed in common malignancies at the transcription levels. High incidences of the breast, lung, and urinary cancers were closely related to the prognostic index for survival. The most prevalent mutation in EIF4A3 was E59K/Q. The tumor necrosis factor-α (TNF-α)/nuclear factor-κB (NF-κB) signaling pathway was affected by these mutations. Co-expression networks showed that EIF4A3 regulates apoptosis and cell cycle via several cancer-related signal pathways, and promotes tumor cell migration, invasion and drug resistance. Conclusion: Our results suggest the potential role for EIF4A3 to serve as a diagnostic marker or therapeutic target for certain types of cancers.
Collapse
Affiliation(s)
- Yan Lin
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Jinyan Zhang
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Junying Cai
- Maternal and Child Health Hospital and Obstetrics and Gynecology Hospital of Guangxi Zhuang Autonomous Region, Guangxi 530003, People's Republic of China
| | - Rong Liang
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Guoying Chen
- The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, People's Republic of China
| | - Gang Qin
- The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, People's Republic of China
| | - Xueqiong Han
- The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, People's Republic of China
| | - Chunling Yuan
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Zhihui Liu
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Yongqiang Li
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Donghua Zou
- The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, People's Republic of China
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|