1
|
Shen Z, Gao Y, Sun X, Chen M, Cen C, Wang M, Wang N, Liu B, Li J, Cui X, Hou J, Shi Y, Gao F. Inactivation of JNK signalling results in polarity loss and cell senescence of Sertoli cell. Cell Prolif 2025; 58:e13760. [PMID: 39329440 PMCID: PMC11839192 DOI: 10.1111/cpr.13760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
As major somatic cells in the testis, Sertoli cell development is precisely regulated by numerous factors, and aberrant development of these cells is associated with male reproductive diseases. JNK signalling is evolutionarily conserved and involved in multiple critical biological processes. Here, we found that the double knockout of Jnk1 and Jnk2 resulted in aberrant localisation of Sertoli cells at early developmental stages, with most Sertoli cells being lost at later stages. Further studies revealed that the inactivation of JNK signalling caused polarity loss in Sertoli cells. In vitro-cultured Jnk1/2-DKO Sertoli cells exhibited a senescence-associated phenotype. Mechanistic studies demonstrate that JNK signalling is likely involved in establishing Sertoli cell polarity by regulating the expression of TGF-β2, mediated by c-Jun. The senescence of Sertoli cells in JNKs-deficient mice is caused by aberrant proteolysis of P27KIP1, mediated by c-Myc. This study demonstrates the role of JNK signalling in Sertoli cell development and functional maintenance, which may also represent an aetiology of male infertility in humans.
Collapse
Affiliation(s)
- Zhiming Shen
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
- Department of Reproductive MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Yang Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xuedong Sun
- Eastern Department of NeurologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Department of NeurologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Changhuo Cen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Mengyue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Nan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Bowen Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiayi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiuhong Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jian Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Yuhua Shi
- Department of Reproductive MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Department of Obstetrics and Gynecology, Center for Reproductive MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
2
|
Wiejak J, Murphy FA, Barker G, Maffia P, Yarwood SJ. Non-cyclic nucleotide EPAC1 activators suppress lipopolysaccharide-regulated gene expression, signalling and intracellular communication in differentiated macrophage-like THP-1 cells. Cell Signal 2024; 124:111444. [PMID: 39368792 DOI: 10.1016/j.cellsig.2024.111444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
This study explores the anti-inflammatory effects of non-cyclic nucleotide EPAC1 activators, PW0577 and SY007, on lipopolysaccharide (LPS)-induced responses in differentiated THP-1 macrophage-like cells. Both activators were found to selectively activate EPAC1 in THP-1 macrophages, leading to the activation of the key down-stream effector, Rap1. RNA sequencing analysis of LPS-stimulated THP-1 macrophages, revealed that treatment with PW0577 or SY007 significantly modulates gene expression related to fibrosis and inflammation, including the suppression of NLRP3, IL-1β, and caspase 1 protein expression in LPS-stimulated cells. Notably, these effects were independent of p65 NFκB phosphorylation at Serine 536, indicating a distinct mechanism of action. The study further identified a shared influence of both activators on LPS signalling pathways, particularly impacting extracellular matrix (ECM) components and NFκB-regulated genes. Additionally, in a co-culture model involving THP-1 macrophages, vascular smooth muscle cells, and human coronary artery endothelial cells, EPAC1 activators modulated immune-vascular interactions, suggesting a broader role in regulating cellular communication between macrophages and endothelial cells. These findings enhance our understanding of EPAC1's role in inflammation and propose EPAC1 activators as potential therapeutic agents for treating inflammatory and fibrotic conditions through targeted modulation of Rap1 and associated signalling pathways.
Collapse
Affiliation(s)
- Jolanta Wiejak
- Heriot-Watt University, Institute of Biological Chemistry, Biophysics and Bioengineering, Edinburgh EH14 4AS, United Kingdom
| | - Fiona A Murphy
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Graeme Barker
- Heriot-Watt University, Institute of Chemical Sciences, Edinburgh EH14 4AS, United Kingdom
| | - Pasquale Maffia
- University of Glasgow, School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, Glasgow G12 8TA, United Kingdom; University of Naples Federico II, Department of Pharmacy, School of Medicine and Surgery, Naples, 80131, Italy; Africa-Europe Cluster of Research Excellence in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance (ARUA) & The Guild of European Research-intensive Universities, Glasgow G12 8TA, United Kingdom
| | - Stephen J Yarwood
- Heriot-Watt University, Institute of Biological Chemistry, Biophysics and Bioengineering, Edinburgh EH14 4AS, United Kingdom.
| |
Collapse
|
3
|
Nnamani PO, Nwagwu C, Diovu EO, Abonyi OE, Nesterkina M, Neu T, Richter C, Loretz B, Lehr CM. Design and evaluation of nanostructured lipid carrier of Bergenin isolated from Pentaclethra macrophylla for anti-inflammatory effect on lipopolysaccharide-induced inflammatory responses in macrophages. Eur J Pharm Biopharm 2024; 200:114307. [PMID: 38685438 DOI: 10.1016/j.ejpb.2024.114307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Herein, we report the properties of nanostructured lipid carriers (NLCs) prepared with a gradient concentration of Bergenin (BGN) isolated from Pentaclethra macrophylla stem bark powder. A gradient concentration of BGN (BGN 0, 50, 100, 150, and 200 mg) was prepared in a 5 % lipid matrix consisting of Transcutol HP (75 %), Phospholipon 90H (15 %), and Gelucire 43/01 (10 %) to which a surfactant aqueous phase consisting of Tween 80, sorbitol, and sorbic acid was dissolved. The NLCs were evaluated by size, polydispersity index (PDI), zeta potential, Fourier Transform Infrared Spectroscopy (FTIR), Differential Scanning Calorimetry (DSC), encapsulation efficiency, and in vitro drug release. The result shows polydispersed nanoparticles with high drug encapsulation (94.26-99.50 %). The nanoparticles were mostly spherical, but those from the 50 mg BGN batch were more cuboidal than spherical. The drug release was highest from the latter to the tune of 40 % compared to the pure BGN solution, which released about 15 % BGN. The anti-inflammatory activity of the BGN-NLC and total plant extract was studied on lipopolysaccharide (LPS)-inflamed macrophages. The cell study showed that BGN and plant extract had low cytotoxicity on macrophages and exhibited a dose-dependent anti-inflammatory effect on the LPS-induced inflammatory process in macrophages.
Collapse
Affiliation(s)
- Petra Obioma Nnamani
- Department of Pharmaceutics, University of Nigeria, Nsukka 40001, Enugu, Nigeria; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany.
| | - Chinekwu Nwagwu
- Department of Pharmaceutics, University of Nigeria, Nsukka 40001, Enugu, Nigeria; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany
| | - Edith Obioma Diovu
- Department of Pharmacognosy and Environmental Medicine, University of Nigeria, Nsukka, Nigeria
| | | | - Mariia Nesterkina
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany
| | - Tobias Neu
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Clémentine Richter
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus Building E 8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
4
|
Kamińska A, Pardyak L, Lustofin S, Gielata K, Arent Z, Pietsch-Fulbiszewska A, Hejmej A. 9-cis-retinoic acid signaling in Sertoli cells regulates their immunomodulatory function to control lymphocyte physiology and Treg differentiation. Reprod Biol Endocrinol 2024; 22:75. [PMID: 38926848 PMCID: PMC11202360 DOI: 10.1186/s12958-024-01246-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Testis is an immune privileged organ, which prevents the immune response against sperm antigens and inflammation. Testicular cells responsible for immune tolerance are mainly Sertoli cells, which form the blood-testis barrier and produce immunosuppressive factors. Sertoli cells prevent inflammation in the testis and maintain immune tolerance by inhibiting proliferation and inducing lymphocyte apoptosis. It has been shown that 9-cis-retinoic acid (9cRA) blocks ex vivo apoptosis of peripheral blood lymphocytes and promotes the differentiation of Treg cells in the gut. However, the role of retinoid signaling in regulating the immune privilege of the testes remains unknown. OBJECTIVE The aim of this study was to determine whether 9cRA, acting via the retinoic acid receptors (RAR) and the retinoic X receptors (RXR), controls the immunomodulatory functions of Sertoli cells by influencing the secretion of anti-inflammatory/pro-inflammatory factors, lymphocyte physiology and Treg cell differentiation. METHODS Experiments were performed using in vitro model of co-cultures of murine Sertoli cells and T lymphocytes. Agonists and antagonists of retinoic acid receptors were used to inhibit/stimulate retinoid signaling in Sertoli cells. RESULTS Our results have demonstrated that 9cRA inhibits the expression of immunosuppressive genes and enhances the expression of pro-inflammatory factors in Sertoli cells and lymphocytes, increases lymphocyte viability and decreases apoptosis rate. Moreover, we have found that 9cRA blocks lymphocyte apoptosis acting through both RAR and RXR and inhibiting FasL/Fas/Caspase 8 and Bax/Bcl-2/Caspase 9 pathways. Finally, we have shown that 9cRA signaling in Sertoli cells inhibits Treg differentiation. CONCLUSION Collectively, our results indicate that retinoid signaling negatively regulates immunologically privileged functions of Sertoli cells, crucial for ensuring male fertility. 9cRA inhibits lymphocyte apoptosis, which can be related to the development of autoimmunity, inflammation, and, in consequence, infertility.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, Krakow, 30-387, Poland.
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University of Agriculture in Kraków, Krakow, 30-248, Poland
| | - Sylwia Lustofin
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, Krakow, 30-387, Poland
| | - Karolina Gielata
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, Krakow, 30-387, Poland
| | - Zbigniew Arent
- Center of Experimental and Innovative Medicine, University of Agriculture in Kraków, Krakow, 30-248, Poland
- University Centre of Veterinary Medicine, University of Agriculture in Kraków, Krakow, 30-059, Poland
| | | | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, Krakow, 30-387, Poland
| |
Collapse
|
5
|
Bahman F, Al-Roub A, Akhter N, Al Madhoun A, Wilson A, Almansour N, Al-Rashed F, Sindhu S, Al-Mulla F, Ahmad R. TNF-α/Stearate Induced H3K9/18 Histone Acetylation Amplifies IL-6 Expression in 3T3-L1 Mouse Adipocytes. Int J Mol Sci 2024; 25:6776. [PMID: 38928498 PMCID: PMC11203872 DOI: 10.3390/ijms25126776] [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: 05/08/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Extensive evidence supports the connection between obesity-induced inflammation and the heightened expression of IL-6 adipose tissues. However, the mechanism underlying the IL-6 exacerbation in the adipose tissue remains unclear. There is general agreement that TNF-α and stearate concentrations are mildly elevated in adipose tissue in the state of obesity. We hypothesize that TNF-α and stearate co-treatment induce the increased expression of IL-6 in mouse adipocytes. We therefore aimed to determine IL-6 gene expression and protein production by TNF-α/stearate treated adipocytes and investigated the mechanism involved. To test our hypothesis, 3T3-L1 mouse preadipocytes were treated with TNF-α, stearate, or TNF-α/stearate. IL-6 gene expression was assessed by quantitative real-time qPCR. IL-6 protein production secreted in the cell culture media was determined by ELISA. Acetylation of histone was analyzed by Western blotting. Il6 region-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac) was determined by ChIP-qPCR. 3T3-L1 mouse preadipocytes were co-challenged with TNF-α and stearate for 24 h, which led to significantly increased IL-6 gene expression (81 ± 2.1 Fold) compared to controls stimulated with either TNF-α (38 ± 0.5 Fold; p = 0.002) or stearate (56 ± 2.0 Fold; p = 0.013). As expected, co-treatment of adipocytes with TNF-α and stearate significantly increased protein production (338 ± 11 pg/mL) compared to controls stimulated with either TNF-α (28 ± 0.60 pg/mL; p = 0.001) or stearate (53 ± 0.20 pg/mL, p = 0.0015). Inhibition of histone acetyltransferases (HATs) with anacardic acid or curcumin significantly reduced the IL-6 gene expression and protein production by adipocytes. Conversely, TSA-induced acetylation substituted the stimulatory effect of TNF-α or stearate in their synergistic interaction for driving IL-6 gene expression and protein production. Mechanistically, TNF-α/stearate co-stimulation increased the promoter-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac), rendering a transcriptionally permissive state that favored IL-6 expression at the transcriptional and translational levels. Our data represent a TNF-α/stearate cooperativity model driving IL-6 expression in 3T3-L1 cells via the H3K9/18Ac-dependent mechanism, with implications for adipose IL-6 exacerbations in obesity.
Collapse
Affiliation(s)
- Fatemah Bahman
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Nadeem Akhter
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Ajit Wilson
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Nourah Almansour
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Fahd Al-Mulla
- Translational Research Department, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.B.); (A.A.-R.); (N.A.); (A.W.); (N.A.); (F.A.-R.); (S.S.)
| |
Collapse
|
6
|
Jazinaki MS, Nosrati M, Chambari M, Jamialahmadi T, Sahebkar A. The Effects of Raspberry Consumption on Glycemic Control and Inflammation Markers in Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr Dev Nutr 2024; 8:102161. [PMID: 38860149 PMCID: PMC11163149 DOI: 10.1016/j.cdnut.2024.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/29/2024] [Accepted: 04/14/2024] [Indexed: 06/12/2024] Open
Abstract
Despite observing the health benefits of raspberry consumption in some recent studies, there is still no consensus regarding this effectiveness on inflammatory markers and glycemic control. This study aimed to investigate this effectiveness by performing a meta-analysis. The PubMed, Web of Science, and Scopus databases were comprehensively searched until December 2023 to find relevant randomized controlled trials. Eligible studies were screened, and relevant information was extracted. The overall effect size of raspberry consumption on each of the outcomes was estimated by following the random-effects model in the form of a 95% confidence interval (CI) and a weighted mean difference (WMD). Raspberry consumption led to a significant increase in insulin concentrations (WMD: 1.89 μU/mL; 95%CI: 1.45, 2.34; P < 0.001) and a significant decrease in tumor necrosis factor-α (TNF-α) concentrations (WMD: -3.07 pg/mL; 95%CI: -5.17, -0.97; P = 0.004), compared with the control groups. Raspberry consumption did not have a significant effect on fasting blood glucose, insulin, hemoglobin A1C, glucose tolerance tests, homeostatic model assessment for insulin resistance, C-reactive protein, and interleukin-6 concentrations. This review revealed that raspberry consumption led to a significant increase and decrease in insulin and TNF-α concentrations, respectively. However, to draw a more accurate conclusion, it is necessary to conduct studies with a larger sample size in the future. The current study's protocol has been registered in the PROSPERO system as CRD42023477559.
Collapse
Affiliation(s)
- Mostafa Shahraki Jazinaki
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Nosrati
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahla Chambari
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Ebrahim Soltani Z, Elahi M, Khavandi M, Haddadi NS, Shayan M, Khalilzadeh M, Dehpour AR. Therapeutic potential of infliximab for pruritus in mice model of cholestasis induced by bile duct ligation: Possible involvement of IL-31. Int Immunopharmacol 2023; 123:110806. [PMID: 37597403 DOI: 10.1016/j.intimp.2023.110806] [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: 06/18/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Cholestatic pruritus is a distressful sensation that can cause a massive desire of scratching skin. Despite maximum medication therapy, some patients still experience pruritus. In this study, we evaluated the effect of infliximab on cholestatic pruritus induced in mice by bile duct ligation. METHODS Twenty-four balb/c mice were randomly assigned to three groups; sham, control, and treatment. The bile duct ligation procedure was performed on mice in the control and treatment groups. After six days, mice in the treatment group received subcutaneous administration of infliximab, and the next day all mice were subjected to the scratching behavior test. Skin, dorsal root ganglia (DRG), and blood samples of mice were collected and evaluated by histopathological, molecular, and biochemical tests. RESULTS The scratching behavior has significantly decreased in mice with cholestasis after the administration of infliximab. The levels of TNFα, TNFR1, TNFR2, NF-κB, and IL-31were higher in control mice compared to sham. In addition, expression levels of TNFR1, NF-κB, and IL-31 were decreased in the treatment group compared to the controls in skin and DRG, while TNFR2 levels were decreased only in DRG. CONCLUSION Infliximab can block TNFα interaction with receptors and inhibit further inflammatory response. Also, our results suggested that infliximab can suppress IL-31 expression indirectly, which is a well-known cytokine in pruritus pathophysiology Infliximab can be a potential therapeutic approach in resistant pruritus in cholestatic disorders.
Collapse
Affiliation(s)
- Zahra Ebrahim Soltani
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Elahi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadmahdi Khavandi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Shayan
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Khalilzadeh
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Wu W, Hu Y, Zhang Q, Xu Y, Su W. TNFα stimulates the proliferation of immature Sertoli cells by attenuating UPS-degradation of cyclin D1 and leads to the delay of BTB maturation in pubertal rats. Andrology 2023; 11:575-590. [PMID: 36354278 DOI: 10.1111/andr.13336] [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: 07/06/2022] [Revised: 09/18/2022] [Accepted: 10/31/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUNDS The Sertoli cell that plays a vital role during spermatogenesis is a known target of physiological and pathological factors affecting testicular development. Tumor necrosis factor alpha (TNFα) participates in the blood-testis barrier reconstruction, cell apoptosis, and inflammatory response by recognizing receptors on Sertoli cell. TNFα has also been shown to induce the proliferation of immature Sertoli cell in vitro, yet the mechanism still remains unclarified. OBJECTIVES This study was designed to investigate the effect of TNFα on blood-testis barrier development during puberty and the underlying mechanisms of TNFα-induced immature Sertoli cell proliferation. MATERIALS AND METHODS Immature male Sprague-Dawley rats of postnatal day 12 were intraperitoneally injected with TNFα. Biotin-labeled method was used to detect permeability of the developing blood-testis barrier after TNFα treatment, and the distribution of occludin and junctional adhesion molecule-A (JAM-A) were detected by immunofluorescence. Sertoli cells isolated from Sprague-Dawley rats of postnatal day 10 were cultured in vitro and treated with TNFα. Cell proliferation rate was reflected by Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assay. Immunoblot and quantitative polymerase chain reaction were used to detect the expression of proliferating cell nuclear antigen, Fbxo4, and cyclin D1. Immunoprecipitation was used to detect the ubiquitination of cyclin D1 and the interaction between Fbxo4 and cyclin D1. Ammonium pyrrolidinedithiocarbamate (PDTC) was applied to detect the effect of nuclear factor kappaB (NFκB) activity inhibition on TNFα-induced Sertoli cell proliferation. The adenoviral recombinant plasmid containing rat Fbxo4 gene was constructed to investigate the effect of Fbxo4 overexpression on Sertoli cell proliferation promoted by TNFα. RESULTS The in vivo experiment revealed a significant delay of blood-testis barrier maturation in pubertal rats caused by exogenous TNFα. TNFα (10 ng/ml) treatment in vitro was found to promote the proliferation of immature Sertoli cells, accompanied with increased NFκB activity and cyclin D1 protein level. The level of Fbxo4 and ubiquitination of cyclin D1 were decreased after TNFα treatment. Inhibitor of NFκB or overexpression of Fbxo4 could both reverse the TNFα-induced proliferation of immature Sertoli cells, meanwhile restore the ubiquitin-proteasome system-dependent degradation of cyclin D1. Overexpression of Fbxo4 could not affect the activation of NFκB caused by TNFα. CONCLUSION These results indicate that TNFα inhibits the ubiquitination and degradation of cyclin D1 through the NFκB pathway, thereby promoting the proliferation of immature Sertoli cell in vitro and inducing the delay of blood-testis barrier maturation in pubertal rats.
Collapse
Affiliation(s)
- Weixing Wu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang, Liaoning, China.,National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Ying Hu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang, Liaoning, China
| | - Qiang Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang, Liaoning, China
| | - Ying Xu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang, Liaoning, China
| | - Wenhui Su
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Saçık U, Çavdar Z, Ural C, Ersoy N, Özoğul C, Erbil G. Effect of ferulic acid on testicular damage caused by torsion-detorsion in rats. Biotech Histochem 2023; 98:77-85. [PMID: 35996879 DOI: 10.1080/10520295.2022.2110615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Testicular torsion is twisting of the spermatic cord around its axis, which impairs blood flow and causes ischemia and formation of free radicals. Ferulic acid is a phenolic acid of the hydroxycinnamic family that is found in the seeds and leaves of plants; it is present in substantial amounts in fruits and vegetables. We investigated the protective effect of ferulic acid on experimental testicular torsion in rats. Animals were divided randomly into five groups: control, ethyl alcohol, torsion, torsion-detorsion, and torsion-detorsion + ferulic acid. Histopathology was assessed using hematoxylin and eosin, and periodic acid-Schiff staining. Tissues were assessed using TUNEL, active caspase-3, myeloperoxidase and inducible nitric oxide synthase immunostaining. Biochemical changes were assessed using assays for superoxide dismutase, malondialdehyde, glutathione peroxidase and glutathione. Ferulic acid reduced the levels of free radicals and increased the levels of antioxidants. Ferulic acid also reduced histopathological changes and germ cell differentiation in the testis following torsion-detorsion. Ferulic acid should be investigated further as a potential treatment for sequelae of torsion-detorsion injury.
Collapse
|
10
|
Ulinastatin ameliorates preeclampsia induced by N(gamma)-nitro-l-arginine methyl ester in a rat model via inhibition of the systemic and placental inflammatory response. J Hypertens 2023; 41:150-158. [PMID: 36453658 DOI: 10.1097/hjh.0000000000003316] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
BACKGROUND The pathogenesis of preeclampsia (PE) is associated with inflammation and endothelial damage. Ulinastatin (UTI) mainly inhibits proteolytic activity and significantly reduces the release of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) from macrophages. It also ameliorates vascular endothelial damage in pathological conditions. Hence, we investigated the effects of UTI in a rat model of PE induced using N(gamma)-nitro-l-arginine methyl ester (L-NAME). METHODS Although inducing PE in a rat model, 5000 U/kg of UTI were injected daily. Systolic blood pressure (SBP) and protein levels in the urine were measured. Renal function, and serum concentrations of TNF-α, IL-6, placental growth factor (PLGF), and von Willebrand factor (vWF) were evaluated. The number and weight of live fetuses as well as the weight of placentas were measured. Placentas were collected for western blot and pathological analysis. RESULTS UTI slightly ameliorated proteinuria and the increases in SBP, blood urea nitrogen (BUN), and serum creatinine. Furthermore, UTI improved serum and placental protein expression levels of TNF-α, IL-6, vWF, and PLGF. Pathological analysis revealed that vascular density and blood flow perfusion was enhanced, vessel wall thickening and neutrophil infiltration were diminished, and the weight and number of live fetuses as well as the weight of the placentas were improved with UTI. CONCLUSION Preventive use of UTI in the PE rat model induced by L-NAME partially alleviated hypertension, proteinuria, and impaired renal function; improved fetal growth restriction; diminished vascular endothelial injury; and ameliorated placental vasculogenesis abnormality and malperfusion by inhibiting the systemic and placental inflammatory response, suggesting that UTI is a potential drug for PE prevention or treatment.
Collapse
|
11
|
Jahan S, Awaja N, Hess B, Hajjar S, Sad S, Lohnes D. The transcription factor Cdx2 regulates inflammasome activity through expression of the NLRP3 suppressor TRIM31 to maintain intestinal homeostasis. J Biol Chem 2022; 298:102386. [PMID: 35985421 PMCID: PMC9508567 DOI: 10.1016/j.jbc.2022.102386] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/03/2022] Open
Abstract
The intestine-specific transcription factor Cdx2 is essential for intestinal homeostasis and has been implicated in the pathogenesis of disorders including inflammatory bowel disease. However, the mechanism by which Cdx2 influences intestinal disease is not clear. Here, we present evidence supporting a novel Cdx2–TRIM31–NLRP3 (NLR family, pyrin domain containing 3) signaling pathway, which may represent a mechanistic means by which Cdx2 impacts intestinal inflammation. We found that conditional loss of Cdx function resulted in an increase in proinflammatory cytokines, including tumor necrosis factor alpha, interleukin (IL)-1β, and IL-6, in the mouse colon. We further show that TRIM31, which encodes a suppressor of NLRP3 (a central component of the NLRP3 inflammasome complex) is a novel Cdx2 target gene and is attenuated in the colon of Cdx conditional mutants. Consistent with this, we found that attenuation of TRIM31 in Cdx mutant intestine occurs concomitant with elevated levels of NLRP3 and an increase in inflammasome products. We demonstrate that specific inhibition of NLRP3 activity significantly reduced IL-1β and IL-6 levels and extended the life span of Cdx conditional mutants, reflecting the therapeutic potential of targeting NLRP3. Tumor necrosis factor-alpha levels were also induced independent of NLRP3, potentially via elevated activity of the proinflammatory NF-κB signaling pathway in Cdx mutants. Finally, in silico analysis of ulcerative colitis patients revealed attenuation of CDX2 and TRIM31 expression coincident with enhanced expression of proinflammatory cytokines. We conclude that the novel Cdx2–TRIM31–NLRP3 signaling pathway promotes proinflammatory cytokine expression, and its inhibition may have therapeutic potential in human intestinal diseases.
Collapse
Affiliation(s)
- Sanzida Jahan
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Nidaa Awaja
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Bradley Hess
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Stephanie Hajjar
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - David Lohnes
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
12
|
Er H, Tas GG, Soygur B, Ozen S, Sati L. Acute and Chronic Exposure to 900 MHz Radio Frequency Radiation Activates p38/JNK-mediated MAPK Pathway in Rat Testis. Reprod Sci 2022; 29:1471-1485. [PMID: 35015292 DOI: 10.1007/s43032-022-00844-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/01/2022] [Indexed: 10/19/2022]
Abstract
The use of electronic devices such as mobile phones has had a long stretch of rapid growth all over the world. Therefore, exposure to radio frequency radiation (RFR) has increased enormously. Here, we aimed to assess the balance between cell death and proliferation and also investigate the involvement of the JNK/p38 MAPK signaling pathway in the testis of rats exposed to 900 MHz RFR in acute and chronic periods (2 h/day, 5 days/week) for 1 or 10 weeks, respectively. The expression of proliferating cell nuclear antigen (PCNA), Bcl-xL, cleaved caspase-3, phosphorylated-JNK (p-JNK), and phosphorylated-p38 (p-p38) was analyzed in line with histopathology and TUNEL analysis in rat testis. There were no histopathological differences between sham and RFR groups in the acute and chronic groups. PCNA expression was not altered between groups in both periods. However, alterations for cleaved caspase-3 and Bcl-xL were observed depending on the exposure period. TUNEL analysis showed a significant increase in the RFR group in the acute period, whereas no difference in the chronic groups for the apoptotic index was reported. In addition, both p-p38 and p-JNK protein expressions increased significantly in RFR groups in both periods. Our study indicated that 900 MHz RFR might result in alterations during acute period exposure for several parameters, but this can be ameliorated in the chronic period in rat testis. Here, we also report the involvement of the p38/JNK-mediated MAPK pathway after exposure to 900 MHz RFR. Hence, this information might shed light in future studies toward detailed molecular mechanisms in male reproduction and infertility.
Collapse
Affiliation(s)
- Hakan Er
- Department of Biophysics, Akdeniz University School of Medicine, Akdeniz University, Antalya, Turkey.,Department of Medical Imaging Techniques, Vocational School of Health Services, Akdeniz University, Antalya, Turkey
| | - Gizem Gamze Tas
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Bikem Soygur
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.,Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Sukru Ozen
- Department of Electrical and Electronics Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
| | - Leyla Sati
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
13
|
Ling S, Ceban F, Lui LMW, Lee Y, Teopiz KM, Rodrigues NB, Lipsitz O, Gill H, Subramaniapillai M, Mansur RB, Lin K, Ho R, Rosenblat JD, Castle D, McIntyre RS. Molecular Mechanisms of Psilocybin and Implications for the Treatment of Depression. CNS Drugs 2022; 36:17-30. [PMID: 34791625 DOI: 10.1007/s40263-021-00877-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/27/2022]
Abstract
Therapeutic deficiencies with monoaminergic antidepressants invites the need to identify and develop novel rapid-acting antidepressants. Hitherto, ketamine and esketamine are identified as safe, well-tolerated rapid-acting antidepressants in adults with treatment-resistant depression, and also mitigate measures of suicidality. Psilocybin is a naturally occurring psychoactive alkaloid and non-selective agonist at many serotonin receptors, especially at serotonin 5-HT2A receptors, and is found in the Psilocybe genus of mushrooms. Preliminary studies with psilocybin have shown therapeutic promise across diverse populations including major depressive disorder. The pharmacodynamic mechanisms mediating the antidepressant and psychedelic effects of psilocybin are currently unknown but are thought to involve the modulation of the serotonergic system, primarily through agonism at the 5-HT2A receptors and downstream changes in gene expression. It is also established that indirect effects on dopaminergic and glutamatergic systems are contributory, as well as effects at other lower affinity targets. Along with the direct effects on neurochemical systems, psilocybin alters neural circuitry and key brain regions previously implicated in depression, including the default mode network and amygdala. The aim of this review is to synthesize the current understanding of the receptor pharmacology and neuronal mechanisms underlying the psychedelic and putative antidepressant properties of psilocybin.
Collapse
Affiliation(s)
- Susan Ling
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Felicia Ceban
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Orly Lipsitz
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Mehala Subramaniapillai
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Kangguang Lin
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University, (Guangzhou Huiai Hospital), Guangzhou Medical University, Guangzhou, China.,Laboratory of Emotion and Cognition, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou Medical University, Guangzhou, China
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - David Castle
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada. .,Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| |
Collapse
|
14
|
Al-Roub A, Al Madhoun A, Akhter N, Thomas R, Miranda L, Jacob T, Al-Ozairi E, Al-Mulla F, Sindhu S, Ahmad R. IL-1β and TNFα Cooperativity in Regulating IL-6 Expression in Adipocytes Depends on CREB Binding and H3K14 Acetylation. Cells 2021; 10:3228. [PMID: 34831450 PMCID: PMC8619559 DOI: 10.3390/cells10113228] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
IL-6 was found to be overexpressed in the adipose tissue of obese individuals, which may cause insulin resistance. However, the regulation of IL-6 in adipocytes in obesity setting remains to be explored. Since IL-1β and TNFα are increased in obese adipose tissue and promote inflammation, we investigated whether cooperation between IL-1β and TNFα influences the production of IL-6. Our data show that IL-1β and TNFα cooperatively enhance IL-6 expression in 3T3L-1 adipocytes. Similar results were seen in human adipocytes isolated from subcutaneous and visceral fat. Although adipocytes isolated from lean and obese adipose tissues showed similar responses for production of IL-6 when incubated with IL-1β/TNFα, secretion of IL-6 was higher in adipocytes from obese tissue. TNFα treatment enhanced CREB binding at CRE locus, which was further enhanced with IL-1β, and was associated with elevated histone acetylation at CRE locus. On the other hand, IL-1β treatments mediated C/EBPβ binding to NF-IL-6 consensus, but not sufficiently to mediate significant histone acetylation. Interestingly, treatment with both stimulatory factors amplifies CREB binding and H3K14 acetylation. Furthermore, histone acetylation inhibition by anacardic acid or curcumin reduces IL-6 production. Notably, inhibition of histone deacetylase (HDAC) activity by trichostatin A (TSA) resulted in the further elevation of IL-6 expression in response to combined treatment of adipocytes with IL-1β and TNFα. In conclusion, our results show that there is an additive interaction between IL-1β and TNFα that depends on CREB binding and H3K14 acetylation, and leads to the elevation of IL-6 expression in adipocytes, providing interesting pathophysiological connection among IL-1β, TNFα, and IL-6 in settings such as obesity.
Collapse
Affiliation(s)
- Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Ashraf Al Madhoun
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Nadeem Akhter
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Lavina Miranda
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Ebaa Al-Ozairi
- Medical Division, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Fahd Al-Mulla
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| |
Collapse
|
15
|
Gao L, Tang Z, Li T, Wang J. Combination of kaempferol and azithromycin attenuates Staphylococcus aureus-induced osteomyelitis via anti-biofilm effects and by inhibiting the phosphorylation of ERK1/2 and SAPK. Pathog Dis 2021; 79:6381690. [PMID: 34610107 DOI: 10.1093/femspd/ftab048] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Osteomyelitis is bacterial infection of bone, commonly caused by Staphylococcus aureus. This work aims to study the potential of azithromycin and kaempferol against chronic osteomyelitis induced by azithromycin-resistant Staphylococcus aureus (ARSA). It was noticed that rats tolerated the treatments with no diarrhoea or weight loss; also, no deaths were observed in rats. The treatment by azithromycin alone failed to inhibit bacterial growth and also had no effect on the infection condition of bone, although the treatment decreased the levels of interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α), but did not improve the oxidative stress levels. Kaempferol monotherapy slightly inhibited bacterial growth and bone infection; the treatment also inhibited the levels of IL-6 and (TNF-α). The treatment also improved the antioxidant status. However, the combined treatment of azithromycin and kaempferol significantly suppressed bacterial growth and bone infection and modulated oxidative stress. In vitro, the combined treatment inhibited the levels of IL-6 and TNF-α, and also suppressed the phosphorylation of ERK1/2 and stress-activated protein kinase (SAPK). The combined treatment also showed anti-biofilm activity in ARSA. The combination attenuates ARSA-induced osteomyelitis in rats compared with their treatments alone by reducing oxidative stress, inhibiting the phosphorylation of ERK1/2 and SAPK and inhibiting biofilm formation.
Collapse
Affiliation(s)
- Lei Gao
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Zhipeng Tang
- Clinical Lab, He Bei General Hospital, Shi Jia Zhuang, 050051, China
| | - Tianbo Li
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Jiangning Wang
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| |
Collapse
|
16
|
Kleandrova VV, Scotti MT, Speck-Planche A. Indirect-Acting Pan-Antivirals vs. Respiratory Viruses: A Fresh Perspective on Computational Multi-Target Drug Discovery. Curr Top Med Chem 2021; 21:2687-2693. [PMID: 34636311 DOI: 10.2174/1568026621666211012110819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022]
Abstract
Respiratory viruses continue to afflict mankind. Among them, pathogens such as coronaviruses [including the current pandemic agent known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] and the one causing influenza A (IAV) are highly contagious and deadly. These can evade the immune system defenses while causing a hyperinflammatory response that can damage different tissues/organs. Simultaneously targeting immunomodulatory proteins is a plausible antiviral strategy since it could lead to the discovery of indirect-acting pan-antiviral (IAPA) agents for the treatment of diseases caused by respiratory viruses. In this context, computational approaches, which are an essential part of the modern drug discovery campaigns, could accelerate the identification of multi-target immunomodulators. This perspective discusses the usefulness of computational multi-target drug discovery for the virtual screening (drug repurposing) of IAPA agents capable of boosting the immune system through the activation of the toll-like receptor 7 (TLR7) and/or the stimulator of interferon genes (STING) while inhibiting key pro-inflammatory proteins, such as caspase-1 and tumor necrosis factor-alpha (TNF-α).
Collapse
Affiliation(s)
- Valeria V Kleandrova
- Laboratory of Fundamental and Applied Research of Quality and Technology of Food Production, Moscow State University of Food Production, Volokolamskoe shosse 11, 125080, Moscow. Russian Federation
| | - Marcus T Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, 58051-900, João Pessoa. Brazil
| | - Alejandro Speck-Planche
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, 58051-900, João Pessoa. Brazil
| |
Collapse
|
17
|
Toricelli M, Evangelista SR, Buck HS, Viel TA. Microdose Lithium Treatment Reduced Inflammatory Factors and Neurodegeneration in Organotypic Hippocampal Culture of Old SAMP-8 Mice. Cell Mol Neurobiol 2021; 41:1509-1520. [PMID: 32642922 PMCID: PMC11448662 DOI: 10.1007/s10571-020-00916-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022]
Abstract
It was already shown that microdoses of lithium carbonate (Li2CO3) promoted memory stabilization in humans and mice. Prolonged treatment also reduced neuronal loss and increased the density of the neurotrophin BDNF in transgenic mice for Alzheimer's disease. The aim of this study was to evaluate whether lithium ions affect inflammatory profiles and neuronal integrity in an animal model of accelerated senescence (SAMP-8). Organotypic hippocampal cultures obtained from 11 to 12-month-old SAMP-8 mice were treated with 2 µM, 20 µM and 200 µM Li2CO3. 2 µM Li2CO3 promoted a significant reduction in propidium iodide uptake in the CA2 area of hippocampus, whereas 20 µM promoted neuroprotection in the CA3 and GrDG areas. 200 µM caused an increase in cellular death, showing toxicity. Measured with quantitative PCR, IL-1α, IL-6 and MIP-1B/CCL-4 gene expression was significantly reduced with 20 µM Li2CO3, whereas IL-10 gene expression was significantly increased with the same concentration. In addition, 2 µM and 20 µM Li2CO3 were also effective in reducing the activation of NFkB and inflammatory cytokines densities, as evaluated by ELISA. It is concluded that very low doses of Li2CO3 can play an important role in neuroprotection as it can reduce neuronal loss and neuroinflammation in older individuals.
Collapse
Affiliation(s)
- Mariana Toricelli
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | | | - Hudson Sousa Buck
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Tania Araujo Viel
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
18
|
Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease. Blood Adv 2021; 5:3445-3456. [PMID: 34438448 DOI: 10.1182/bloodadvances.2020004016] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Idiopathic multicentric Castleman disease (iMCD) is a poorly understood hematologic disorder involving cytokine-induced polyclonal lymphoproliferation, systemic inflammation, and potentially fatal multiorgan failure. Although the etiology of iMCD is unknown, interleukin-6 (IL-6) is an established disease driver in approximately one-third of patients. Anti-IL-6 therapy, siltuximab, is the only US Food and Drug Administration-approved treatment. Few options exist for siltuximab nonresponders, and no validated tests are available to predict likelihood of response. We procured and analyzed the largest-to-date cohort of iMCD samples, which enabled classification of iMCD into disease categories, discovery of siltuximab response biomarkers, and identification of therapeutic targets for siltuximab nonresponders. Proteomic quantification of 1178 analytes was performed on serum of 88 iMCD patients, 60 patients with clinico-pathologically overlapping diseases (human herpesvirus-8-associated MCD, N = 20; Hodgkin lymphoma, N = 20; rheumatoid arthritis, N = 20), and 42 healthy controls. Unsupervised clustering revealed iMCD patients have heterogeneous serum proteomes that did not cluster with clinico-pathologically overlapping diseases. Clustering of iMCD patients identified a novel subgroup with superior response to siltuximab, which was validated using a 7-analyte panel (apolipoprotein E, amphiregulin, serum amyloid P-component, inactivated complement C3b, immunoglobulin E, IL-6, erythropoietin) in an independent cohort. Enrichment analyses and immunohistochemistry identified Janus kinase (JAK)/signal transducer and activator of transcription 3 signaling as a candidate therapeutic target that could potentially be targeted with JAK inhibitors in siltuximab nonresponders. Our discoveries demonstrate the potential for accelerating discoveries for rare diseases through multistakeholder collaboration.
Collapse
|
19
|
Kočović DM, Bajuk-Bogdanović D, Pećinar I, Nedeljković BB, Daković M, Andjus PR. Assessment of cellular and molecular changes in the rat brain after gamma radiation and radioprotection by anisomycin. JOURNAL OF RADIATION RESEARCH 2021; 62:793-803. [PMID: 34062561 PMCID: PMC8438266 DOI: 10.1093/jrr/rrab045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/31/2021] [Indexed: 06/12/2023]
Abstract
The objective of the study was to describe cellular and molecular markers of radioprotection by anisomycin, focusing on the changes in rat brain tissue. Two-month-old Wistar rats were exposed to a 60Co radiation source at a dose of 6 Gy, with or without radioprotection with anisomycin (150 mg/kg) administered subcutaneously 30 min before or 3 or 6 h after irradiation. Survivors were analyzed 30 days after treatment. Astroglial and microglial responses were investigated based on the expression of glial markers assessed with immunohistochemistry, and quantitative changes in brain biomolecules were investigated by Raman microspectroscopy. In addition, blood plasma levels of pro-inflammatory (interleukin 6 and tumor necrosis factor α) and anti-inflammatory (interleukin 10) cytokines were assessed. We found that application of anisomycin either before or after irradiation significantly decreased the expression of the microglial marker Iba-1. We also found an increased intensity of Raman spectral bands related to nucleic acids, as well as an increased level of cytokines when anisomycin was applied after irradiation. This suggests that the radioprotective effects of anisomycin are by decreasing Iba-1 expression and stabilizing genetic material by increasing the level of nucleic acids.
Collapse
Affiliation(s)
- Dušica M Kočović
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Danica Bajuk-Bogdanović
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Ilinka Pećinar
- Faculty of Agriculture, Department for Agrobotany, University of Belgrade, Nemanjina 6, 11 080 Belgrade, Serbia
| | - Biljana Božić Nedeljković
- Institute for Physiology and Biochemistry ``Jean Giaja'', Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Marko Daković
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Pavle R Andjus
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| |
Collapse
|
20
|
Hsu CC, Li Y, Hsu CT, Cheng JT, Lin MH, Cheng KC, Chen SW. Etanercept Ameliorates Cardiac Fibrosis in Rats with Diet-Induced Obesity. Pharmaceuticals (Basel) 2021; 14:320. [PMID: 33916242 PMCID: PMC8067047 DOI: 10.3390/ph14040320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diet-induced obesity (DIO) is considered the main risk factor for cardiovascular diseases. Increases in the plasma levels of tumor necrosis factor alpha (TNF-α) is associated with DIO. Etanercept, a TNF-α inhibitor, has been shown to alleviate cardiac hypertrophy. To investigate the effect of etanercept on cardiac fibrosis in DIO model, rats on high fat diet (HFD) were subdivided into two groups: the etanercept group and vehicle group. Cardiac injury was identified by classic methods, while fibrosis was characterized by histological analysis of the hearts. Etanercept treatment at 0.8 mg/kg/week twice weekly by subcutaneous injection effectively alleviates the cardiac fibrosis in HFD-fed rats. STAT3 activation seems to be induced in parallel with fibrosis-related gene expression in the hearts of HFD-fed rats. Decreased STAT3 activation plays a role in the etanercept-treated animals. Moreover, fibrosis-related genes are activated by palmitate in parallel with STAT3 activation in H9c2 cells. Etanercept may inhibit the effects of palmitate, but it is less effective than a direct inhibitor of STAT3. Direct inhibition of STAT3 activation by etanercept seems unlikely. Etanercept has the ability to ameliorate cardiac fibrosis through reduction of STAT3 activation after the inhibition of TNF-α and/or its receptor.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department of Exercise and Health Sciences, University of Taipei, Taipei City 110, Taiwan;
- Department of Otorhinolaryngology, Taipei City Hospital, Taipei City 110, Taiwan
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Guishan, Taoyuan City 613, Taiwan;
| | - Yingxiao Li
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 970, Taiwan;
| | - Chao-Tien Hsu
- Department of Pathology, I-Shou University Medical Center, Yanchao District, Kaohsiung City 824, Taiwan;
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Tainan City 710, Taiwan;
| | - Mang-Hung Lin
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Guishan, Taoyuan City 613, Taiwan;
| | - Kai-Chun Cheng
- Department of Pharmacy, College of Pharmacy, Tajen University, Pingtung 907, Taiwan
| | - Shang-Wen Chen
- Division of Cardiology, Chi-Mei Medical Center Liouying, Tainan City 736, Taiwan
| |
Collapse
|
21
|
Spence T, Allsopp PJ, Yeates AJ, Mulhern MS, Strain JJ, McSorley EM. Maternal Serum Cytokine Concentrations in Healthy Pregnancy and Preeclampsia. J Pregnancy 2021; 2021:6649608. [PMID: 33680514 PMCID: PMC7925069 DOI: 10.1155/2021/6649608] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
The maternal immune response is essential for successful pregnancy, promoting immune tolerance to the fetus while maintaining innate and adaptive immunity. Uncontrolled, increased proinflammatory responses are a contributing factor to the pathogenesis of preeclampsia. The Th1/Th2 cytokine shift theory, characterised by bias production of Th2 anti-inflammatory cytokine midgestation, was frequently used to reflect the maternal immune response in pregnancy. This theory is simplistic as it is based on limited information and does not consider the role of other T cell subsets, Th17 and Tregs. A range of maternal peripheral cytokines have been measured in pregnancy cohorts, albeit the changes in individual cytokine concentrations across gestation is not well summarised. Using available data, this review was aimed at summarising changes in individual maternal serum cytokine concentrations throughout healthy pregnancy and evaluating their association with preeclampsia. We report that TNF-α increases as pregnancy progresses, IL-8 decreases in the second trimester, and IL-4 concentrations remain consistent throughout gestation. Lower second trimester IL-10 concentrations may be an early predictor for developing preeclampsia. Proinflammatory cytokines (TNF-α, IFN-γ, IL-2, IL-8, and IL-6) are significantly elevated in preeclampsia. More research is required to determine the usefulness of using cytokines, particularly IL-10, as early biomarkers of pregnancy health.
Collapse
Affiliation(s)
- Toni Spence
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Philip J. Allsopp
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Alison J. Yeates
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Maria S. Mulhern
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - J. J. Strain
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Emeir M. McSorley
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
22
|
Di Persio S, Starace D, Capponi C, Saracino R, Fera S, Filippini A, Vicini E. TNF-α inhibits GDNF levels in Sertoli cells, through a NF-κB-dependent, HES1-dependent mechanism. Andrology 2021; 9:956-964. [PMID: 33314792 DOI: 10.1111/andr.12959] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/13/2020] [Accepted: 12/08/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Glial cell line-derived neurotrophic factor (GDNF) is a soluble molecule crucial for the regulation of the spermatogonial stem cells (SSC) of the testis. The effects of GDNF on target cells have been extensively described, but mechanisms underlying GDNF regulation are currently under investigation. In the nervous system, GDNF expression is regulated by pro-inflammatory cytokines including lipopolysaccharide (LPS), interleukin 1 beta (IL-1β), and tumor necrosis factor alpha (TNF-α) but the effect of these cytokines on GDNF expression in the testis is unclear. OBJECTIVES The aim of the present study was to investigate the impact of TNF-α on GDNF expression levels using primary murine Sertoli cells as experimental model. MATERIAL AND METHODS The expression of TNF-α-regulated genes including Gdnf in different culture conditions was determined by real-time PCR. GDNF protein levels were determined by ELISA. The activation of the NF-κb pathway and HES1 levels were assessed by Western Blot analysis and immunofluorescence. HES1 expression was downregulated by RNAi. RESULTS In primary Sertoli cells, TNF-α downregulates GDNF levels through a nuclear factor-κB (NF-κB)-dependent mechanism. Mechanistically, TNF-α induces the transcriptional repressor HES1 by a NF-Κb-dependent mechanism, which in turn downregulates GDNF. DISCUSSION Under physiological conditions, TNF-α is secreted by germ cells suggesting that this cytokine plays a role in the paracrine control of SSC niche by modulating GDNF levels. HES1, a well-known target of the Notch pathway, is implicated in the regulation of GDNF expression. In Sertoli cells, TNF-α and Notch signaling may converge at molecular level, to regulate the expression of HES1 and HES1- target genes, including GDNF. CONCLUSIONS Because of the importance of GDNF for spermatogonial stem cell self-renewal and proliferation, this data may give important insights on how cytokine signals in the testis modulate the expression of niche-derived factors.
Collapse
Affiliation(s)
- Sara Di Persio
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Donatella Starace
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Chiara Capponi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Rossana Saracino
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Stefania Fera
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| | - Elena Vicini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
23
|
Zhang C. Flare-up of cytokines in rheumatoid arthritis and their role in triggering depression: Shared common function and their possible applications in treatment (Review). Biomed Rep 2020; 14:16. [PMID: 33269077 PMCID: PMC7694594 DOI: 10.3892/br.2020.1392] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/31/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic illnesses are associated with an increased risk of depression and anxiety. Rheumatoid arthritis (RA) is a chronic autoimmune disease that typically causes damage to the joints. RA extensively impacts patients, both physically and psychologically. Depression is a common comorbid disorder with RA, which leads to worsened health outcomes. There are several cytokines that are active in the joints of patients with RA. Inflammatory cytokines serve important roles in the key processes in the joints, which usually cause inflammation, articular damage and other comorbidities associated with RA. The key role of inflammatory cytokines could be attributed to their interactions within signaling pathways. In RA, IL-1, and the cytokines of TNF-α, IL-6 and IL-18 are primarily involved. Furthermore, depression is hypothesized to be strongly associated with systemic inflammation, particularly with dysregulation of the cytokine network. The present review summarizes the current state of knowledge on these two diseases from the perspective of inflammation and cytokines, and emphasizes the possible bridge between them by exploring the involvement of systemic cytokines in both conditions.
Collapse
Affiliation(s)
- Chunhai Zhang
- Thyroid Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin 1300332, P.R. China
| |
Collapse
|
24
|
Adhikari A, Cobb B, Eddington S, Becerra N, Kohli P, Pond A, Davie J. IFN-γ and CIITA modulate IL-6 expression in skeletal muscle. Cytokine X 2020; 2:100023. [PMID: 33604554 PMCID: PMC7885875 DOI: 10.1016/j.cytox.2020.100023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/20/2022] Open
Abstract
Interleukin 6 (IL-6) is a secreted cytokine that is an important mediator of the immune response in numerous tissues, including skeletal muscle. IL-6 is considered a myokine as it can be secreted by muscle. IL-6 is secreted following exercise, where it exerts both pro-myogenic effects as well as anti-myogenic effects such as promoting atrophy and muscle wasting. The regulation of IL-6 in skeletal muscle is not well understood. The purpose of this study was to determine if IFN-γ and TNF-ɑ stimulate IL-6 in skeletal muscle. We found that both IFN-γ and TNF-α stimulate IL-6 in skeletal muscle, but the stimulation is not cooperative as seen in monocytes. We have previously shown that the IFN-γ stimulated class II major histocompatibility complex transactivator (CIITA) mediates many of the effects of IFN-γ in skeletal muscle and we show here that CIITA directly stimulates IL-6. The regulation of IL-6 by CIITA is clearly complex, as we found that CIITA both stimulates and restrains IL-6 expression. To show that these effects could be observed in a physiological setting, mice were treated with IFN-γ and we found that both CIITA and IL-6 were upregulated in skeletal muscle.
Collapse
Affiliation(s)
- Abhinav Adhikari
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Brittan Cobb
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Seth Eddington
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Nathalie Becerra
- Department of Chemistry, Southern Illinois Carbondale, Carbondale, IL, USA
| | - Punit Kohli
- Department of Chemistry, Southern Illinois Carbondale, Carbondale, IL, USA
| | - Amber Pond
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Judith Davie
- Department of Biochemistry and Molecular Biology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| |
Collapse
|
25
|
Ibrahim RR, Amer RA, Abozeid AA, Elsharaby RM, Shafik NM. Micro RNA 146a gene variant / TNF-α / IL-6 / IL-1 β; A cross-link axis inbetween oxidative stress, endothelial dysfunction and neuro-inflammation in acute ischemic stroke and chronic schizophrenic patients. Arch Biochem Biophys 2020; 679:108193. [DOI: 10.1016/j.abb.2019.108193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
26
|
Fu MH, Chen IC, Lee CH, Wu CW, Lee YC, Kung YC, Hung CY, Wu KLH. Anti-neuroinflammation ameliorates systemic inflammation-induced mitochondrial DNA impairment in the nucleus of the solitary tract and cardiovascular reflex dysfunction. J Neuroinflammation 2019; 16:224. [PMID: 31729994 PMCID: PMC6858639 DOI: 10.1186/s12974-019-1623-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/24/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Decreased heart rate variability (HRV) leads to cardiovascular diseases and increased mortality in clinical studies. However, the underlying mechanisms are still inconclusive. Systemic inflammation-induced neuroinflammation is known to impair the autonomic center of cardiovascular regulation. The dynamic stability of blood pressure and heart rate (HR) is regulated by modulation of the reciprocal responses of sympathetic and parasympathetic tone by the baroreflex, which is controlled by the nucleus of the solitary tract (NTS). METHODS Systemic inflammation was induced by E. coli lipopolysaccharide (LPS, 1.2 mg/kg/day, 7 days) peritoneal infusion via an osmotic minipump in normotensive Sprague-Dawley rats. Systolic blood pressure (SBP) and HR were measured by femoral artery cannulation and recorded on a polygraph under anesthesia. The low-frequency (LF; 0.25-0.8 Hz) and high-frequency (HF; 0.8-2.4 Hz) components of SBP were adopted as the indices for sympathetic vasomotor tone and parasympathetic vasomotor tone, while the baroreflex effectiveness index (BEI) was adopted from the analysis of SBP and pulse interval (PI). The plasma levels of proinflammatory cytokines and mitochondrial DNA (mtDNA) oxidative damage were analyzed by ELISA. Protein expression was evaluated by Western blot. The distribution of oxidative mtDNA was probed by immunofluorescence. Pharmacological agents were delivered via infusion into the cisterna magna with an osmotic minipump. RESULTS The suppression of baroreflex sensitivity was concurrent with increased SBP and decreased HR. Neuroinflammatory factors, including TNF-α, CD11b, and Iba-1, were detected in the NTS of the LPS group. Moreover, indices of mtDNA damage, including 8-OHdG and γ-H2AX, were significantly increased in neuronal mitochondria. Pentoxifylline or minocycline intracisternal (IC) infusion effectively prevented mtDNA damage, suggesting that cytokine and microglial activation contributed to mtDNA damage. Synchronically, baroreflex sensitivity was effectively protected, and the elevated blood pressure was significantly relieved. In addition, the mtDNA repair mechanism was significantly enhanced by pentoxifylline or minocycline. CONCLUSION These results suggest that neuronal mtDNA damage in the NTS induced by neuroinflammation could be the core factor in deteriorating baroreflex desensitization and subsequent cardiovascular dysfunction. Therefore, the enhancement of base excision repair (BER) signaling in mitochondria could be a potential therapeutic strategy for cardiovascular reflex dysregulation.
Collapse
Affiliation(s)
- Mu-Hui Fu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan Republic of China
| | - I-Chun Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Chou-Hwei Lee
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Chih-Wei Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Yu-Chi Lee
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Yu Chih Kung
- Master of Science Program in Health Care, Department of Nursing, Meiho University, Neipu Township, Republic of China
- Department of Nursing, Meiho University, Neipu Township, Taiwan, Republic of China
| | - Chun-Ying Hung
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
| | - Kay L. H. Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301 Taiwan Republic of China
- Department of Senior Citizen Services, National Tainan Institute of Nursing, Tainan, 700 Taiwan Republic of China
| |
Collapse
|
27
|
Ni FD, Hao SL, Yang WX. Multiple signaling pathways in Sertoli cells: recent findings in spermatogenesis. Cell Death Dis 2019; 10:541. [PMID: 31316051 PMCID: PMC6637205 DOI: 10.1038/s41419-019-1782-z] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/25/2022]
Abstract
The functions of Sertoli cells in spermatogenesis have attracted much more attention recently. Normal spermatogenesis depends on Sertoli cells, mainly due to their influence on nutrient supply, maintenance of cell junctions, and support for germ cells' mitosis and meiosis. Accumulating evidence in the past decade has highlighted the dominant functions of the MAPK, AMPK, and TGF-β/Smad signaling pathways during spermatogenesis. Among these pathways, the MAPK signaling pathway regulates dynamics of tight junctions and adherens junctions, proliferation and meiosis of germ cells, proliferation and lactate production of Sertoli cells; the AMPK and the TGF-β/Smad signaling pathways both affect dynamics of tight junctions and adherens junctions, as well as the proliferation of Sertoli cells. The AMPK signaling pathway also regulates lactate supply. These signaling pathways combine to form a complex regulatory network for spermatogenesis. In testicular tumors or infertile patients, the activities of these signaling pathways in Sertoli cells are abnormal. Clarifying the mechanisms of signaling pathways in Sertoli cells on spermatogenesis provides new insights into the physiological functions of Sertoli cells in male reproduction, and also serves as a pre-requisite to identify potential therapeutic targets in abnormal spermatogenesis including testicular tumor and male infertility.
Collapse
Affiliation(s)
- Fei-Da Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Shuang-Li Hao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
Johnson SL, Kirk RD, DaSilva NA, Ma H, Seeram NP, Bertin MJ. Polyphenol Microbial Metabolites Exhibit Gut and Blood⁻Brain Barrier Permeability and Protect Murine Microglia against LPS-Induced Inflammation. Metabolites 2019; 9:metabo9040078. [PMID: 31010159 PMCID: PMC6523162 DOI: 10.3390/metabo9040078] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence supports the beneficial effects of polyphenol-rich diets, including the traditional Mediterranean diet, for the management of cardiovascular disease, obesity and neurodegenerative diseases. However, a common concern when discussing the protective effects of polyphenol-rich diets against diseases is whether these compounds are present in systemic circulation in their intact/parent forms in order to exert their beneficial effects in vivo. Here, we explore two common classes of dietary polyphenols, namely isoflavones and lignans, and their gut microbial-derived metabolites for gut and blood-brain barrier predicted permeability, as well as protection against neuroinflammatory stimuli in murine BV-2 microglia. Polyphenol microbial metabolites (PMMs) generally showed greater permeability through artificial gut and blood-brain barriers compared to their parent compounds. The parent polyphenols and their corresponding PMMs were evaluated for protective effects against lipopolysaccharide-induced inflammation in BV-2 microglia. The lignan-derived PMMs, equol and enterolactone, exhibited protective effects against nitric oxide production, as well as against pro-inflammatory cytokines (IL-6 and TNF-α) in BV-2 microglia. Therefore, PMMs may contribute, in large part, to the beneficial effects attributed to polyphenol-rich diets, further supporting the important role of gut microbiota in human health and disease prevention.
Collapse
Affiliation(s)
- Shelby L Johnson
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Riley D Kirk
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Nicholas A DaSilva
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Hang Ma
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Navindra P Seeram
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Matthew J Bertin
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
29
|
Grosicka-Maciąg E, Kurpios-Piec D, Woźniak K, Kowalewski C, Szumiło M, Drela N, Kiernozek E, Suchocki P, Rahden-Staroń I. Selol (Se IV) modulates adhesive molecules in control and TNF-α-stimulated HMEC-1 cells. J Trace Elem Med Biol 2019; 51:106-114. [PMID: 30466918 DOI: 10.1016/j.jtemb.2018.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
Selol, an organic selenitetrigliceride formulation containing selenium at +4 oxidation level, has been suggested as anticancer drug. One of the causes of several diseases including cancer may be inflammation. This study aimed at determining the activity of Selol via measuring its effect on reactive oxygen species (ROS) generation, nuclear factor kappa B (NF-κB) activation, intercellular cell adhesion molecules-1 (ICAM-1), vascular cell adhesive molecule-1 (VCAM-1), and plateled-endothelial cell adhesive molecule-1 (PECAM-1) levels on control and on tumor necrosis factor-α (TNF-α)-stimulated human microvascular endothelial cells (HMEC-1). Cells were treated either with Selol 5% (4 or 8 μgSe/mL) or TNF-α (10 ng/mL) alone or with Selol concomitant with TNF-α. Selol treatment resulted in ROS generation, activation of NF-κB, downregulation of PECAM-1, VCAM-1 and slight upregulation ICAM-1 expression on the cell surface. TNF-α treatment reflected in sharp NF-κB activation, upregulation of both ICAM-1 and VCAM-1 in parallel with the downregulation of PECAM-1 expression on cell surface. Exposure to both compounds upregulated ICAM-1 and VCAM-1, downregulated PECAM-1 level on cell surface in parallel with no changes in level of NF-κB activation as compared with effects mediated by TNF-α alone. These results points to new look at Selol action since it shows a pro-inflammatory activity in parallel with effects on CAMs expression on the cell surface of human microvascular endothelial cells. However, since Selol enhances CAMs expression level when is present concomitantly with TNF-α this fact might suggest that selenium present in the condition of inflammation will make it worse.
Collapse
Affiliation(s)
- Emilia Grosicka-Maciąg
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warszawa, Banacha 1, Poland.
| | - Dagmara Kurpios-Piec
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warszawa, Banacha 1, Poland.
| | - Katarzyna Woźniak
- Department of Dermatology and Immunodermatology, Medical University of Warsaw, 02-008 Warszawa, Koszykowa 82a, Poland.
| | - Cezary Kowalewski
- Department of Dermatology and Immunodermatology, Medical University of Warsaw, 02-008 Warszawa, Koszykowa 82a, Poland.
| | - Maria Szumiło
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warszawa, Banacha 1, Poland.
| | - Nadzieja Drela
- Immunology Department, Faculty of Biology, University of Warsaw, 02-096 Warszawa, Miecznikowa 1, Poland.
| | - Ewelina Kiernozek
- Immunology Department, Faculty of Biology, University of Warsaw, 02-096 Warszawa, Miecznikowa 1, Poland.
| | - Piotr Suchocki
- Department of Bioanalysis and Drug Analysis, Medical University of Warsaw, 02-097 Warszawa, Banacha 1, Poland.
| | - Iwonna Rahden-Staroń
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warszawa, Banacha 1, Poland.
| |
Collapse
|
30
|
Rutting S, Xenaki D, Malouf M, Horvat JC, Wood LG, Hansbro PM, Oliver BG. Short-chain fatty acids increase TNFα-induced inflammation in primary human lung mesenchymal cells through the activation of p38 MAPK. Am J Physiol Lung Cell Mol Physiol 2018; 316:L157-L174. [PMID: 30407866 DOI: 10.1152/ajplung.00306.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs), produced as by-products of dietary fiber metabolism by gut bacteria, have anti-inflammatory properties and could potentially be used for the treatment of inflammatory diseases, including asthma. The direct effects of SCFAs on inflammatory responses in primary human lung mesenchymal cells have not been assessed. We investigated whether SCFAs can protect against tumor necrosis factor (TNF)α-induced inflammation in primary human lung fibroblasts (HLFs) and airway smooth muscle (ASM) cells in vitro. HLFs and ASM cells were exposed to SCFAs, acetate (C2:0), propionate (C3:0), and butyrate (C4:0) (0.01-25 mM) with or without TNFα, and the release of proinflammatory cytokines, IL-6, and CXCL8 was measured using ELISA. We found that none of the SCFAs suppressed TNFα-induced cytokine release. On the contrary, challenge with supraphysiological concentrations (10-25 mM), as might be used therapeutically, of propionate or butyrate in combination with TNFα resulted in substantially greater IL-6 and CXCL8 release from HLFs and ASM cells than challenge with TNFα alone, demonstrating synergistic effects. In ASM cells, challenge with acetate also enhanced TNFα-induced IL-6, but not CXCL8 release. Synergistic upregulation of IL-6 and CXCL8 was mediated through the activation of free fatty acid receptor (FFAR)3, but not FFAR2. The signaling pathways involved were further examined using specific inhibitors and immunoblotting, and responses were found to be mediated through p38 MAPK signaling. This study demonstrates that proinflammatory, rather than anti-inflammatory effects of SCFAs are evident in lung mesenchymal cells.
Collapse
Affiliation(s)
- Sandra Rutting
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Dia Xenaki
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia
| | - Monique Malouf
- Thoracic Medicine and Lung Transplantation, Saint Vincent's Hospital , Sydney, New South Wales , Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia.,Graduate School of Health, Discipline of Pharmacy, University of Technology Sydney , Sydney, New South Wales , Australia
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia.,School of Life Sciences, University of Technology Sydney , Sydney, New South Wales , Australia
| |
Collapse
|
31
|
Luca G, Arato I, Sorci G, Cameron DF, Hansen BC, Baroni T, Donato R, White DGJ, Calafiore R. Sertoli cells for cell transplantation: pre-clinical studies and future perspectives. Andrology 2018; 6:385-395. [DOI: 10.1111/andr.12484] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/08/2023]
Affiliation(s)
- G. Luca
- Department of Experimental Medicine; University of Perugia; Perugia Italy
- Division of Medical Andrology and Endocrinology of Reproduction; University of Perugia and Saint Mary Hospital; Terni Italy
| | - I. Arato
- Department of Experimental Medicine; University of Perugia; Perugia Italy
| | - G. Sorci
- Department of Experimental Medicine; University of Perugia; Perugia Italy
- Inter-University Institute of Myology (IIM)
| | - D. F. Cameron
- Department of Pathology and Cell Biology; Morsani College of Medicine; University of South Florida; Tampa FL USA
| | - B. C. Hansen
- Department of Internal Medicine and Pediatrics; Morsani College of Medicine; University of South Florida; Tampa FL USA
| | - T. Baroni
- Department of Experimental Medicine; University of Perugia; Perugia Italy
| | - R. Donato
- Department of Experimental Medicine; University of Perugia; Perugia Italy
- Inter-University Institute of Myology (IIM)
- Centro Universitario per la Ricerca sulla Genomica Funzionale; Perugia Italy
| | - D. G. J. White
- Robarts Research Institute; University of Western Ontario; London ON Canada
| | - R. Calafiore
- Division of Medical Andrology and Endocrinology of Reproduction; University of Perugia and Saint Mary Hospital; Terni Italy
- Department of Medicine; University of Perugia; Perugia Italy
| |
Collapse
|
32
|
Rutting S, Xenaki D, Lau E, Horvat J, Wood LG, Hansbro PM, Oliver BG. Dietary omega-6, but not omega-3, polyunsaturated or saturated fatty acids increase inflammation in primary lung mesenchymal cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L922-L935. [PMID: 29368548 DOI: 10.1152/ajplung.00438.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Obesity is an important risk factor for developing severe asthma. Dietary fatty acids, which are increased in sera of obese individuals and after high-fat meals, activate the innate immune system and induce inflammation. This study investigated whether dietary fatty acids directly cause inflammation and/or synergize with obesity-induced cytokines in primary human pulmonary fibroblasts in vitro. Fibroblasts were challenged with BSA-conjugated fatty acids [ω-6 polyunsaturated fatty acids (PUFAs) and ω-3 PUFAs or saturated fatty acids (SFAs)], with or without TNF-α, and release of the proinflammatory cytokines, IL-6 and CXCL8, was measured. We found that the ω-6 PUFA arachidonic acid (AA), but not ω-3 PUFAs or SFAs, upregulates IL-6 and CXCL8 release. Combined AA and TNF-α challenge resulted in substantially greater cytokine release than either alone, demonstrating synergy. Synergistic upregulation of IL-6, but not CXCL8, was mainly mediated via cyclooxygenase (COX). Inhibition of p38 MAPK reduced CXCL8 release, induced by AA and TNF-α alone, but not in combination. Synergistic CXCL8 release, following AA and TNF-α challenge, was not medicated via a single signaling pathway (MEK1, JNK, phosphoinositide 3-kinase, and NF-κB) nor by hyperactivation of NF-κB or p38. To investigate if these findings occur in other airway cells, effects of AA in primary human airway smooth muscle (ASM) cells and human bronchial epithelial cells were also investigated. We found proinflammatory effects in ASM cells but not epithelial cells. This study suggests that diets rich in ω-6 PUFAs might promote airway inflammation via multiple pathways, including COX-dependent and -independent pathways, and in an obese person, may lead to more severe airway inflammation.
Collapse
Affiliation(s)
- Sandra Rutting
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia.,Department of Respiratory Medicine, Royal Prince Alfred Hospital, University of Sydney, Sydney, Australia
| | - Dia Xenaki
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia
| | - Edmund Lau
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, University of Sydney, Sydney, Australia
| | - Jay Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia.,School of Life Sciences, University of Technology Sydney , Sydney , Australia
| |
Collapse
|
33
|
Gudowska-Sawczuk M, Wrona A, Gruszewska E, Cylwik B, Panasiuk A, Flisiak R, Chrostek L. Serum level of interleukin-6 (IL-6) and N-terminal propeptide of procollagen type I (PINP) in patients with liver diseases. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 78:125-130. [DOI: 10.1080/00365513.2017.1420217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | - Alicja Wrona
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Gruszewska
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Bogdan Cylwik
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Anatol Panasiuk
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, Bialystok, Poland
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, Bialystok, Poland
| | - Lech Chrostek
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
34
|
Li W, Cui N, Mazzuca MQ, Mata KM, Khalil RA. Increased vascular and uteroplacental matrix metalloproteinase-1 and -7 levels and collagen type I deposition in hypertension in pregnancy: role of TNF-α. Am J Physiol Heart Circ Physiol 2017. [PMID: 28626073 PMCID: PMC5625170 DOI: 10.1152/ajpheart.00207.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preeclampsia is a pregnancy-related disorder manifested as maternal hypertension in pregnancy (HTN-Preg) and fetal growth restriction. Placental ischemia could be an initiating event that leads to abnormal vascular and uteroplacental remodeling in HTN-Preg; however, the molecular targets and intermediary mechanisms involved are unclear. We tested the hypothesis that placental ischemia could target vascular and uteroplacental matrix metalloproteinases (MMPs) through an inflammatory cytokine-mediated mechanism. MMP levels and distribution were measured in the aorta, uterus, and placenta of normal pregnant (Preg) rats and pregnant rats with reduced uterine perfusion pressure (RUPP). Maternal blood pressure was higher and the litter size and pup weight were lower in RUPP compared with Preg rats. Gelatin zymography showed prominent uterine MMP-2 and MMP-9 activity that was dependent on the amount of loaded protein. At saturating protein loading, both gelatin and casein zymography revealed two additional bands corresponding to MMP-1 and MMP-7 that were greater in the aorta, uterus, and placenta of RUPP compared with Preg rats. Western blots and immunohistochemistry confirmed increased MMP-1 and MMP-7 in the aorta, uterus, and placenta of RUPP versus Preg rats. The levels of MMP-1 and MMP-7 substrate collagen type I were greater in tissues of RUPP compared with Preg rats. In organ culture, TNF-α increased MMP-1 and MMP-7 in the aorta, uterus, and placenta of Preg rats, and a TNF-α antagonist prevented the increases in MMPs in tissues of RUPP rats. Thus, placental ischemia, possibly through TNF-α, increases vascular and uteroplacental MMP-1 and MMP-7, which, in turn, alter collagen deposition and cause inadequate tissue remodeling in HTN-Preg. Cytokine antagonists may reverse the increase in MMP-1 and MMP-7 expression/activity and, in turn, restore proper vascular and uteroplacental remodeling in HTN-Preg and preeclampsia.NEW & NOTEWORTHY The molecular mechanisms of preeclampsia are unclear, making it difficult to predict, prevent, or manage the pregnancy-associated disorder. This study showed that placental ischemia, possibly through the release of TNF-α, causes increases in the levels of matrix metalloproteinase (MMP)-1 and MMP-7, which could alter collagen deposition and cause inadequate uteroplacental and vascular remodeling in hypertension in pregnancy. The data suggest that targeting MMP-1 and MMP-7 and their upstream modulators, such as TNF-α, could provide a new approach in the management of hypertension in pregnancy and preeclampsia.
Collapse
Affiliation(s)
- Wei Li
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ning Cui
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marc Q Mazzuca
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Karina M Mata
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Havunen R, Siurala M, Sorsa S, Grönberg-Vähä-Koskela S, Behr M, Tähtinen S, Santos JM, Karell P, Rusanen J, Nettelbeck DM, Ehrhardt A, Kanerva A, Hemminki A. Oncolytic Adenoviruses Armed with Tumor Necrosis Factor Alpha and Interleukin-2 Enable Successful Adoptive Cell Therapy. MOLECULAR THERAPY-ONCOLYTICS 2016; 4:77-86. [PMID: 28345026 PMCID: PMC5363700 DOI: 10.1016/j.omto.2016.12.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/20/2016] [Indexed: 01/18/2023]
Abstract
Adoptive cell therapy holds much promise in the treatment of cancer but results in solid tumors have been modest. The notable exception is tumor-infiltrating lymphocyte (TIL) therapy of melanoma, but this approach only works with high-dose preconditioning chemotherapy and systemic interleukin (IL)-2 postconditioning, both of which are associated with toxicities. To improve and broaden the applicability of adoptive cell transfer, we constructed oncolytic adenoviruses coding for human IL-2 (hIL2), tumor necrosis factor alpha (TNF-α), or both. The viruses showed potent antitumor efficacy against human tumors in immunocompromised severe combined immunodeficiency (SCID) mice. In immunocompetent Syrian hamsters, we combined the viruses with TIL transfer and were able to cure 100% of the animals. Cured animals were protected against tumor re-challenge, indicating a memory response. Arming with IL-2 and TNF-α increased the frequency of both CD4+ and CD8+ TILs in vivo and augmented splenocyte proliferation ex vivo, suggesting that the cytokines were important for T cell persistence and proliferation. Cytokine expression was limited to tumors and treatment-related signs of systemic toxicity were absent, suggesting safety. To conclude, cytokine-armed oncolytic adenoviruses enhanced adoptive cell therapy by favorable alteration of the tumor microenvironment. A clinical trial is in progress to study the utility of Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123) in human patients with cancer.
Collapse
Affiliation(s)
- Riikka Havunen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | | | - Michael Behr
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Siri Tähtinen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland
| | - Pauliina Karell
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland
| | - Juuso Rusanen
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | | | - Anja Ehrhardt
- Faculty of Health, Institute for Virology and Microbiology, University Witten/Herdecke, 58448 Witten, Germany
| | - Anna Kanerva
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Central Hospital, 00610 Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; TILT Biotherapeutics, Ltd., 00290 Helsinki, Finland; Comprehensive Cancer Center, Helsinki University Hospital, 00290 Helsinki, Finland
| |
Collapse
|
36
|
Roh SG, Suzuki Y, Gotoh T, Tatsumi R, Katoh K. Physiological Roles of Adipokines, Hepatokines, and Myokines in Ruminants. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 29:1-15. [PMID: 26732322 PMCID: PMC4698675 DOI: 10.5713/ajas.16.0001r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Since the discovery of leptin secreted from adipocytes, specialized tissues and cells have been found that secrete the several peptides (or cytokines) that are characterized to negatively and positively regulate the metabolic process. Different types of adipokines, hepatokines, and myokines, which act as cytokines, are secreted from adipose, liver, and muscle tissue, respectively, and have been identified and examined for their physiological roles in humans and disease in animal models. Recently, various studies of these cytokines have been conducted in ruminants, including dairy cattle, beef cattle, sheep, and goat. Interestingly, a few cytokines from these tissues in ruminants play an important role in the post-parturition, lactation, and fattening (marbling) periods. Thus, understanding these hormones is important for improving nutritional management in dairy cows and beef cattle. However, to our knowledge, there have been no reviews of the characteristics of these cytokines in beef and dairy products in ruminants. In particular, lipid and glucose metabolism in adipose tissue, liver tissue, and muscle tissue are very important for energy storage, production, and synthesis, which are regulated by these cytokines in ruminant production. In this review, we summarize the physiological roles of adipokines, hepatokines, and myokines in ruminants. This discussion provides a foundation for understanding the role of cytokines in animal production of ruminants.
Collapse
Affiliation(s)
- Sang-Gun Roh
- Kuju Agriculture Research Center, Kyushu University, Oita 878-020, Japan
| | - Yutaka Suzuki
- Kuju Agriculture Research Center, Kyushu University, Oita 878-020, Japan
| | - Takafumi Gotoh
- Kuju Agriculture Research Center, Kyushu University, Oita 878-020, Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | - Kazuo Katoh
- Kuju Agriculture Research Center, Kyushu University, Oita 878-020, Japan
| |
Collapse
|
37
|
Xu B, Yang H, Sun M, Chen H, Jiang L, Zheng X, Ding G, Liu Y, Sheng Y, Cui D, Duan Y. 2,3',4,4',5-Pentachlorobiphenyl Induces Inflammatory Responses in the Thyroid Through JNK and Aryl Hydrocarbon Receptor-Mediated Pathway. Toxicol Sci 2015; 149:300-11. [PMID: 26519956 DOI: 10.1093/toxsci/kfv235] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are durable and widely distributed environmental contaminants that can compromise the normal functions of multiple organs and systems; one important mechanism is the induction of inflammatory disorders. In this study, we explored the influences of 2,3',4,4',5-pentachlorobiphenyl (PCB118) on inflammatory responses and its underlying mechanisms in the thyroid. Wistar rats were administered PCB118 intraperitoneally at 0, 10, 100, and 1000 μg/kg/d, 5 days a week for 13 weeks; rat thyroid FRTL-5 cells were treated with PCB118 (0, 0.25, 2.5, and 25 nM) for indicated time. Results revealed that PCB118 promoted the generation of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and intercellular adhesion molecule-1 (ICAM-1) in a time- and dose-related manner and decreased sodium/iodide symporter (NIS) protein expression. Moreover, stimulation with PCB118 resulted in the upregulation of the aryl hydrocarbon receptor (AhR)-responsive gene cytochrome P450 1A1 in FRTL-5 cells; whereas pretreatment with the AhR inhibitor α-naphthoflavone or AhR small interfering RNA (siRNA) suppressed AhR, CYP1A1, IL-6, and ICAM-1 and restored NIS expression. In vivo and in vitro studies also suggested that the c-Jun N-terminal kinase (JNK) pathway was activated on PCB118 exposure, and the experiments using siRNA for JNK partially blocked PCB118-induced upregulation of IL-6 and ICAM-1 and downregulation of NIS. Altogether, PCB118 stimulates production of IL-6, TNF-α, and ICAM-1 in the thyroid through AhR and JNK activations and subsequently interferes with NIS expression, resulting in the disruption of thyroid structure and function.
Collapse
Affiliation(s)
- Bojin Xu
- *Department of Endocrinology and
| | - Hui Yang
- *Department of Endocrinology and
| | | | | | | | | | | | - Yun Liu
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yunlu Sheng
- Department of Gerontology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dai Cui
- *Department of Endocrinology and
| | - Yu Duan
- *Department of Endocrinology and
| |
Collapse
|
38
|
Dogan C, Halici Z, Topcu A, Cadirci E, Karakus E, Bayir Y, Selli J. Effects of amlodipine on ischaemia/reperfusion injury in the rat testis. Andrologia 2015; 48:441-52. [PMID: 26259852 DOI: 10.1111/and.12464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2015] [Indexed: 12/27/2022] Open
Abstract
The aim of this study was to examine the effects of amlodipine (AML) in rat testicular torsion/detorsion damage. In this study, rats were divided into eight groups: (i) sham; (ii) testicular ischaemia, 2 h of ischaemia; (iii) testicular ischaemia/reperfusion (I/R), 2 h of ischaemia followed by 2 h of reperfusion; (iv) ischaemia + AML (5 mg kg(-1)) administered 30 min before ischaemia; (v) ischaemia + AML (10 mg kg(-1)) administered 30 min before ischaemia; (vi) and (vii) I/R + AML (5 mg kg(-1)) and I/R + AML (10 mg kg(-1)) administered 1.5 h after the induction of ischaemia, respectively, and at the end of a 2-h ischaemia period and a 2-h reperfusion period applied; and (viii) sham + AML (10 mg kg(-1)). Significant decreases in levels of superoxide dismutase and glutathione were observed in ischaemia and reperfusion groups when compared with healthy controls. These antioxidant levels increased in AML groups while malondialdehyde levels significantly decreased. While increases in tumour necrosis factor-alpha and transforming growth factor-beta levels were found in the torsion and detorsion groups, significant decreases in the levels of these inflammatory cytokines were observed in the treatment groups. These results demonstrate that AML significantly produced protective effects on testis tissue damage that occurs in the torsion/detorsion model via biochemical, histopathological and molecular pathways.
Collapse
Affiliation(s)
- C Dogan
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Z Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - A Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - E Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - E Karakus
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Y Bayir
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - J Selli
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
39
|
|
40
|
Lydka M, Bilinska B, Cheng CY, Mruk DD. Tumor necrosis factor α-mediated restructuring of the Sertoli cell barrier in vitro involves matrix metalloprotease 9 (MMP9), membrane-bound intercellular adhesion molecule-1 (ICAM-1) and the actin cytoskeleton. SPERMATOGENESIS 2014; 2:294-303. [PMID: 23248771 PMCID: PMC3521752 DOI: 10.4161/spmg.22602] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The mammalian blood-testis barrier (BTB) restructures throughout spermatogenesis, thereby allowing developing germ cells to enter the adluminal compartment of the seminiferous epithelium. Previous studies have shown pro-inflammatory cytokines such as tumor necrosis factor α (TNFα) and interleukin-1α to be important regulators of Sertoli cell barrier/BTB function in vitro and in vivo. In this study, the effects of TNFα on Sertoli cell barrier function were assessed, with emphasis on changes in proteases and cell adhesion molecules following treatment. By immunoblotting and immunohistochemistry, MMP9 was found to be present in germ cells, localizing by and large to spermatocytes and spermatids in the adult rat testis. Following treatment of Sertoli cells with physiologically relevant consecutive doses of recombinant human TNFα (25 ng/ml), the steady-state levels of active-matrix metalloprotease 9 (MMP9), membrane-bound intercellular adhesion molecule (mICAM-1) and androgen receptor increased significantly. TNFα also downregulated the steady-state level of occludin, in agreement with earlier results that showed TNFα to disrupt Sertoli cell barrier/BTB function. In addition, TNFα affected the filamentous actin cytoskeleton in Sertoli cells, which appeared to be mediated by cortactin, a regulator of actin dynamics. Taken collectively, these findings imply that germ cells may be involved in BTB restructuring via the localized production of TNFα. These results also illustrate that barrier restructuring correlated with an increase in Sertoli cell mICAM-1, suggesting that it may be critical for adhesion as germ cells traverse the “opened” BTB.
Collapse
Affiliation(s)
- Marta Lydka
- Center for Biomedical Research; Population Council; New York, NY USA
| | | | | | | |
Collapse
|
41
|
Bojalil R, Mata-González MT, Sánchez-Muñoz F, Yee Y, Argueta I, Bolaños L, Amezcua-Guerra LM, Camacho-Villegas TA, Sánchez-Castrejón E, García-Ubbelohde WJ, Licea-Navarro AF, Márquez-Velasco R, Paniagua-Solís JF. Anti-tumor necrosis factor VNAR single domains reduce lethality and regulate underlying inflammatory response in a murine model of endotoxic shock. BMC Immunol 2013; 14:17. [PMID: 23548047 PMCID: PMC3621089 DOI: 10.1186/1471-2172-14-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 03/11/2013] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND In sepsis, tumor necrosis factor (TNF) is the key factor triggering respiratory burst, tissue injury and disseminated coagulation. Anti-TNF strategies based on monoclonal antibodies or F(ab')₂ fragments have been used in sepsis with contradictory results. Immunoglobulin new antigen receptors (IgNAR) are a unique subset of antibodies consisting of five constant (CNAR) and one variable domains (VNAR). VNAR domains are the smallest, naturally occurring, antibody-based immune recognition units, having potential use as therapy. Our aim was to explore the impact of an anti-TNF VNAR on survival in an experimental model of endotoxic shock. Also, mRNA expression and serum protein of several inflammatory molecules were measured. RESULTS Endotoxic shock was induced by lipopolysaccharide (LPS) in male Balb/c mice. Animals were treated with anti-TNF VNAR domains, F(ab')₂ antibody fragments, or saline solution 15 minutes before, 2 h and 24 h after lethal dose₁₀₀ (LD₁₀₀) LPS administration. TNF blockade with either VNAR domains or F(ab')₂ fragments were associated with lower mortality (60% and 75%, respectively) compared to LD₁₀₀. Challenge with LPS induced significant production of serum TNF and interleukins -10 and -6 at 3 h. After that, significant reduction of IL-6 at 24 h (vs 3 h) was shown only in the VNAR group. Nitrites level also increased in response to LPS. In liver, TNF and IL-10 mRNA expression showed a pro-inflammatory imbalance in response to LPS. Blocking TNF was associated with a shift towards an anti-inflammatory status; however, polarization was more pronounced in animals receiving F(ab')₂ fragments than in those with VNAR therapy. With regard to IL-6, gene expression was increased at 3 h in all groups. TNF blockade was associated with rapid and sustained suppression of IL-6 expression, even more evident in the VNAR group. Finally, expression of inducible-nitric oxide synthase (iNOS) increased in response to LPS at 3 h, but this was decreased at 24 h only in the anti-TNF VNAR group. CONCLUSIONS Anti-TNF VNAR single domains improved survival in a murine model of endotoxic shock. Protection was associated with regulation in the TNF/IL-10 balance, attenuation of IL-6 and iNOS gene expression in the liver as well as decreased serum IL-6 concentration.
Collapse
Affiliation(s)
- Rafael Bojalil
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Xiao X, Mruk DD, Cheng CY. Intercellular adhesion molecules (ICAMs) and spermatogenesis. Hum Reprod Update 2013; 19:167-86. [PMID: 23287428 DOI: 10.1093/humupd/dms049] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND During the seminiferous epithelial cycle, restructuring takes places at the Sertoli-Sertoli and Sertoli-germ cell interface to accommodate spermatogonia/spermatogonial stem cell renewal via mitosis, cell cycle progression and meiosis, spermiogenesis and spermiation since developing germ cells, in particular spermatids, move 'up and down' the seminiferous epithelium. Furthermore, preleptotene spermatocytes differentiated from type B spermatogonia residing at the basal compartment must traverse the blood-testis barrier (BTB) to enter the adluminal compartment to prepare for meiosis at Stage VIII of the epithelial cycle, a process also accompanied by the release of sperm at spermiation. These cellular events that take place at the opposite ends of the epithelium are co-ordinated by a functional axis designated the apical ectoplasmic specialization (ES)-BTB-basement membrane. However, the regulatory molecules that co-ordinate cellular events in this axis are not known. METHODS Literature was searched at http://www.pubmed.org and http://scholar.google.com to identify published findings regarding intercellular adhesion molecules (ICAMs) and the regulation of this axis. RESULTS Members of the ICAM family, namely ICAM-1 and ICAM-2, and the biologically active soluble ICAM-1 (sICAM-1) are the likely regulatory molecules that co-ordinate these events. sICAM-1 and ICAM-1 have antagonistic effects on the Sertoli cell tight junction-permeability barrier, involved in Sertoli cell BTB restructuring, whereas ICAM-2 is restricted to the apical ES, regulating spermatid adhesion during the epithelial cycle. Studies in other epithelia/endothelia on the role of the ICAM family in regulating cell movement are discussed and this information has been evaluated and integrated into studies of these proteins in the testis to create a hypothetical model, depicting how ICAMs regulate junction restructuring events during spermatogenesis. CONCLUSIONS ICAMs are crucial regulatory molecules of spermatogenesis. The proposed hypothetical model serves as a framework in designing functional experiments for future studies.
Collapse
Affiliation(s)
- Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
43
|
Nan Y, Zhang X, Yang G, Xie J, Lu Z, Wang W, Ni X, Cao X, Ma J, Wang Z. Icariin stimulates the proliferation of rat Sertoli cells in an ERK1/2-dependent manner in vitro. Andrologia 2012; 46:9-16. [PMID: 23134192 DOI: 10.1111/and.12035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2012] [Indexed: 12/01/2022] Open
Abstract
Icariin (ICA), a major constituent of flavonoids from the Chinese medical herb Epimedium brevicornum Maxim, is found to be protective for male reproductive ability, with the underlying mechanism largely unknown. Our study here investigated the effects of ICA on Sertoli cells, which act as nurse cells for the germ cells developing. Icariin was found to stimulate Sertoli cell proliferation in a dose-dependent manner. Further study revealed that Icariin induced an obvious phosphorylation of ERK in Sertoli cells. Inhibition of activation of ERK by the ERK inhibitor U0126 nearly blocked the Icariin-induced proliferation of Sertoli cells. Taken together, our results suggest that Icariin promotes the proliferation of Sertoli cells in vitro by activating the ERK1/2 signal pathway, which might at least partially, explain the protective role of Icariin on male reproductive ability.
Collapse
Affiliation(s)
- Y Nan
- Department of Chinese Medicine, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| | - X Zhang
- Department of Chinese Medicine, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| | - G Yang
- Department of Biochemistry, the Fourth Military Medical University, Xi'an, China
| | - J Xie
- Department of Chinese Medicine, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| | - Z Lu
- Department of Biochemistry, the Fourth Military Medical University, Xi'an, China
| | - W Wang
- Center of Infectious Diseases, the Fourth Military Medical University, Xi'an, China
| | - X Ni
- Department of Tanesthesia, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| | - X Cao
- Department of Pathology, the Fourth Military Medical University, Xi'an, China
| | - J Ma
- Department of Chinese Medicine, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| | - Z Wang
- Department of Chinese Medicine, Xijing Hospital affiliated to the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
44
|
Xiao X, Cheng CY, Mruk DD. Intercellular adhesion molecule-1 is a regulator of blood-testis barrier function. J Cell Sci 2012; 125:5677-89. [PMID: 22976294 DOI: 10.1242/jcs.107987] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mechanism underlying the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier (BTB) during spermatogenesis is not well understood largely owing to the fact that the BTB, unlike most other blood-tissue barriers, is composed of several co-existing and co-functioning junction types. In the present study, we show that intercellular adhesion molecule-1 [ICAM-1, a Sertoli and germ cell adhesion protein having five immunoglobulin (Ig)-like domains, in addition to transmembrane and cytoplasmic domains] is a regulator of BTB integrity. Initial experiments showed ICAM-1 to co-immunoprecipitate and co-localize with tight junction and basal ectoplasmic specialization proteins such as occludin and N-cadherin, which contribute to BTB function. More importantly, overexpression of ICAM-1 in Sertoli cells in vitro enhanced barrier function when monitored by transepithelial electrical resistance measurements, illustrating that ICAM-1-mediated adhesion can promote BTB integrity. On the other hand, overexpression of a truncated form of ICAM-1 that consisted only of the five Ig-like domains (sICAM-1; this form of ICAM-1 is known to be secreted) elicited an opposite effect when Sertoli cell barrier function was found to be perturbed in vitro; in this case, sICAM-1 overexpression resulted in the downregulation of several BTB constituent proteins, which was probably mediated by Pyk2/p-Pyk2-Y402 and c-Src/p-Src-Y530. These findings were expanded to the in vivo level when BTB function was found to be disrupted following sICAM-1 overexpression. These data illustrate the existence of a unique mechanism in the mammalian testis where ICAM-1 can either positively or negatively regulate BTB function.
Collapse
Affiliation(s)
- Xiang Xiao
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
45
|
Nuclear factor-κB1 controls the functional maturation of dendritic cells and prevents the activation of autoreactive T cells. Nat Med 2011; 17:1663-7. [PMID: 22081022 DOI: 10.1038/nm.2556] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 10/12/2011] [Indexed: 01/08/2023]
Abstract
Mature dendritic cells (DCs) are crucial for the induction of adaptive immune responses and perturbed DC homeostasis can result in autoimmune disease. Either uncontrolled expansion or enhanced survival of DCs can result in a variety of autoimmune diseases in mouse models. In addition, increased maturation signals, through overexpression of surface Toll-like receptors (TLRs) or stimulation by type I interferon (IFN), has been associated with systemic autoimmunity. Whereas recent studies have focused on identifying factors required for initiating the maturation process, the possibility that resting DCs also express molecules that 'hold' them in an immature state has generally not been considered. Here we show that nuclear factor-κB1 (NF-κB1) is crucial for maintaining the resting state of DCs. Self-antigen-pulsed unstimulated DCs that do not express NF-κB1 were able to activate CD8(+) T lymphocytes and induce autoimmunity. We further show that NF-κB1 negatively regulates the spontaneous production of tumor necrosis factor-α (TNF-α), which is associated with increased granzyme B expression in cytotoxic T lymphocytes (CTLs). These findings provide a new perspective on functional DC maturation and a potential mechanism that may account for pathologic T cell activation.
Collapse
|
46
|
Kazutaka S, Winnall WR, Muir JA, Hedger MP. Regulation of Sertoli cell activin A and inhibin B by tumour necrosis factor α and interleukin 1α: interaction with follicle-stimulating hormone/adenosine 3',5'-cyclic phosphate signalling. Mol Cell Endocrinol 2011; 335:195-203. [PMID: 21256182 DOI: 10.1016/j.mce.2011.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 01/13/2011] [Accepted: 01/14/2011] [Indexed: 10/18/2022]
Abstract
Regulation of crucial events during spermatogenesis involves dynamic changes in cytokine production and interactions across the cycle of the seminiferous epithelium. Regulation of activin A and inhibin B production by the inflammatory cytokines, tumour necrosis factor α (TNFα) and interleukin 1α (IL1α), alone and in conjunction with FSH or a cAMP analogue (dibutyryl cAMP), was examined in cultures of Sertoli cells from 20-day old rats. Both TNFα and IL1α stimulated activin A secretion and expression of its subunit (β(A)) mRNA, and suppressed inhibin B secretion and expression of its subunit (α and β(B)) mRNAs. The actions of TNFα and IL1α were opposed by FSH and dibutyryl cAMP. Both cytokines inhibited FSH/dibutyryl cAMP-stimulated inhibin B secretion and mRNA expression as well as stem cell factor mRNA expression. Both cytokines also inhibited FSH-induced cAMP production, and reduced baseline FSH receptor mRNA expression. These data highlight the reciprocal relationship that exists between FSH/cAMP signalling and inflammatory cytokine signalling pathways in the control of Sertoli cell function, and production of activin A/inhibin B in particular. It is anticipated that these interactions play important roles in the fine control of events during the cycle of the seminiferous epithelium and in the inhibition of spermatogenesis during inflammation.
Collapse
Affiliation(s)
- Saito Kazutaka
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
47
|
Hedger MP. Toll-like receptors and signalling in spermatogenesis and testicular responses to inflammation--a perspective. J Reprod Immunol 2011; 88:130-41. [PMID: 21333360 PMCID: PMC7127151 DOI: 10.1016/j.jri.2011.01.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 01/16/2011] [Accepted: 01/19/2011] [Indexed: 02/06/2023]
Abstract
It is self-evident that infection and inflammation in the reproductive tract can inhibit male fertility, but the observation that fertility may also be compromised by systemic inflammation and disease is more difficult to explain. Recent studies implicating microbial pattern-recognition receptors, such as the Toll-like receptors (TLRs), as well as inflammatory cytokines and their signalling pathways, in testicular function have cast new light on this mysterious link between infection/inflammation and testicular dysfunction. It is increasingly evident that signalling pathways normally involved in controlling inflammation play fundamental roles in regulating Sertoli cell activity and responses to reproductive hormones, in addition to promoting immune responses within the testis. Many of the negative effects of inflammation on spermatogenesis may be attributed to elevated production of inflammation-related gene products within the circulation and the testis, which subsequently exert disruptive effects on spermatogenic cell development and survival, as well as the ability of the Sertoli cells to provide support for spermatogenesis. These interactions have important implications for testicular dysfunction and disease, and may eventually provide new opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Mark P Hedger
- Centre for Reproduction and Development, Monash Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton, Melbourne, Victoria 3168, Australia.
| |
Collapse
|
48
|
Turpeinen T, Nieminen R, Moilanen E, Korhonen R. Mitogen-activated protein kinase phosphatase-1 negatively regulates the expression of interleukin-6, interleukin-8, and cyclooxygenase-2 in A549 human lung epithelial cells. J Pharmacol Exp Ther 2010; 333:310-8. [PMID: 20089808 DOI: 10.1124/jpet.109.157438] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mitogen-activated protein kinase phosphatase (MKP)-1 is a protein phosphatase that regulates the activity of p38 mitogen-activated protein (MAP) kinase and c-Jun NH2-terminal kinase (JNK) and, to lesser extent, p42/44 extracellular signal-regulated kinase. Studies with MKP-1(-/-) mice show that MKP-1 is a regulating factor suppressing excessive cytokine production and inflammatory response. The data on the role of MKP-1 in the regulation of inflammatory gene expression in human cells are much more limited. In the present study, we investigated the effect of MKP-1 on the expression of interleukin (IL)-6, IL-8 and cyclooxygenase (COX)-2 in response to stimulation with cytokines (tumor necrosis factor, IL-1beta, and interferon-gamma; 10 ng/ml each) in A549 human lung epithelial cells. Cytokines enhanced p38 and JNK phosphorylation and MKP-1 expression. p38 MAP kinase inhibitors 4-[4-(4-fluorophenyl)-5-(4-pyridinyl)-1H-imidazol-2-yl] phenol (SB202190) and 1-(5-tert-butyl-2-p-tolyl-2H-pyrazol-3-yl)-3(4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl)urea (BIRB 796) inhibited cytokine-induced phosphorylation of p38 substrate MAP kinase-activated protein kinase 2 and expression of IL-6, IL-8, and COX-2. An aminopyridine-based JNK inhibitor, N-(4-amino-5-cyano-6-ethoxypyridin-2-yl)-2-(2,5-dimethoxyphenyl)acetamide (JNK inhibitor VIII), inhibited phosphorylation of a JNK substrate c-Jun but did not have any effect on IL-6, IL-8, or COX-2 expression. Down-regulation of MKP-1 with small interfering RNA enhanced p38 and JNK phosphorylation and increased IL-6, IL-8, and COX-2 expression in A549 cells. In conclusion, cytokine-induced MKP-1 expression was found to negatively regulate p38 phosphorylation and the expression of IL-6, IL-8, and COX-2 in human pulmonary epithelial cells. Our results suggest that MKP-1 is an important negative regulator of inflammatory gene expression in human pulmonary epithelial cells, and compounds that enhance MKP-1 may have anti-inflammatory effects and control inflammatory response in the human lung.
Collapse
Affiliation(s)
- Tuija Turpeinen
- The Immunopharmacology Research Group, Medical School, University of Tampere, FIN-33014 Tampere, Finland
| | | | | | | |
Collapse
|
49
|
Stevens AJ, Gahan ME, Mahalingam S, Keller PA. The medicinal chemistry of dengue fever. J Med Chem 2010; 52:7911-26. [PMID: 19739651 DOI: 10.1021/jm900652e] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Andrew J Stevens
- Department of Chemistry, University of Wollongong, Wollongong 2522, Australia
| | | | | | | |
Collapse
|
50
|
Abstract
A large body of evidence points to the existence of a close, dynamic relationship between the immune system and the male reproductive tract, which has important implications for our understanding of both systems. The testis and the male reproductive tract provide an environment that protects the otherwise highly immunogenic spermatogenic cells and sperm from immunological attack. At the same time, secretions of the testis, including androgens, influence the development and mature functions of the immune system. Activation of the immune system has negative effects on both androgen and sperm production, so that systemic or local infection and inflammation compromise male fertility. The mechanisms underlying these interactions have begun to receive the attention from reproductive biologists and immunologists that they deserve, but many crucial details remain to be uncovered. A complete picture of male reproductive tract function and its response to toxic agents is contingent upon continued exploration of these interactions and the mechanisms involved.
Collapse
Key Words
- cytokines
- immunity
- immunoregulation
- inflammation
- leydig cell
- lymphocytes
- macrophages
- nitric oxide
- prostanoids
- seminal plasma
- sertoli cell
- sperm
- spermatogenesis
- steroidogenesis
- toll-like receptors
- 16:0a-lpc, 1-palmitoyl-sn-glycero-3-phosphocholine
- 18:1a-lpc, 1-oleoyl-sn-glycero-3-phosphocholine
- 18:2a-lpc, 1-linoleoyl-sn-glycero-3-phosphocholine
- 20:4a-lpc, 1-arachidonyl-sn-glycero-3-phosphocholine
- aid, acquired immune deviation
- aire, autoimmune regulator
- ap1, activated protein 1
- apc, antigen-presenting cell
- bambi, bmp and activin membrane-bound inhibitor
- bmp, bone morphogenetic protein
- cox, cyclooxygenase
- crry, complement receptor-related protein
- ctl, cytotoxic t lymphocyte
- eao, experimental autoimmune orchitis
- eds, ethane dimethane sulfonate
- enos, endothelial nos
- fadd, fas-associated death domain protein
- fasl, fas ligand
- fsh, follicle-stimulating hormone
- gc, glucocorticoid
- hcg, human chorionic gonadotropin
- hla, human leukocyte antigen
- hmgb1, high mobility group box chromosomal protein 1
- ice, il1 converting enzyme
- ifn, interferon
- ifnar, ifnα receptor
- il, interleukin
- il1r, interleukin 1 receptor
- il1ra, il1 receptor antagonist
- inos, inducible nitric oxide synthase
- irf, interferon regulatory factor
- jak/stat, janus kinase/signal transducers and activators of transcription
- jnk, jun n-terminal kinase
- lh, luteinizing hormone
- lpc, lysoglycerophosphatidylcholine
- lps, lipopolysaccharide
- map, mitogen-activated protein
- mhc, major histocompatibility complex
- mif, macrophage migration inhibitory factor
- myd88, myeloid differentiation primary response protein 88
- nfκb, nuclear factor kappa b
- nk, cell natural killer cell
- nkt cell, natural killer t cell
- nlr, nod-like receptor
- nnos, neuronal nos
- nod, nucleotide binding oligomerization domain
- p450c17, 17α-hydroxylase/c17-c20 lyase
- p450scc, cholesterol side-chain cleavage complex
- paf, platelet-activating factor
- pamp, pathogen-associated molecular pattern
- pc, phosphocholine
- pg, prostaglandin
- pges, pge synthase
- pgi, prostacyclin
- pla2, phospholipase a2
- pmn, polymorphonuclear phagocyte
- pparγ, peroxisome proliferator-activated receptor γ
- rig, retinoic acid-inducible gene
- rlh, rig-like helicase
- ros, reactive oxygen species
- star, steroidogenic acute regulatory
- tcr, t cell receptor
- tgf, transforming growth factor
- th cell, helper t cell
- tir, toll/il1r
- tlr, toll-like receptor
- tnf, tumor necrosis factor
- tnfr, tnf receptor
- tr1, t regulatory 1
- tradd, tnfr-associated death domain protein
- traf, tumor necrosis factor receptor-associated factor
- treg, regulatory t cell
- trif, tir domain-containing adaptor protein inducing interferon β
- tx, thromboxane
- txas, thromboxane a synthase
Collapse
|