1
|
Zhen Y, Li X, Huang S, Wang R, Yang L, Huang Y, Yan J, Ju J, Wen H, Sun Q. LncRNA lnc-SPRR2G-2 contributes to keratinocyte hyperproliferation and inflammation in psoriasis by activating the STAT3 pathway and downregulating KHSRP. Mol Cell Probes 2024; 76:101967. [PMID: 38942130 DOI: 10.1016/j.mcp.2024.101967] [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: 02/22/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Psoriasis is a chronic inflammatory disease characterized by increased keratinocyte proliferation and local inflammation. Long noncoding RNAs (lncRNAs) play important regulatory roles in many immune-mediated diseases, including psoriasis. In this study, we aimed to investigate the role and mechanism of lnc-SPRR2G-2 (SPRR2G) in M5-treated psoriatic keratinocytes. Fluorescence in situ hybridization and quantitative real-time polymerase chain reaction (qRT-PCR) showed that lnc-SPRR2G-2 was significantly upregulated in psoriasis tissues and psoriatic keratinocytes. In psoriatic keratinocytes, functional and molecular experiment analyses demonstrated that SPRR2G regulated proliferation, cell cycle and apoptosis, and induced the expression of S100 calcium binding protein A7 (S100A7), interleukin (IL)-1β, IL-8 and C-X-C motif chemokine ligand 10 (CXCL10). The function of SPRR2G in psoriasis is related to the STAT3 signaling pathway and can be inhibited by a STAT3 inhibitor. Moreover, KH-type splicing regulatory protein (KHSRP) was proved to be regulated by lnc-SPRR2G-2 and to control the mRNA decay of psoriasis-related cytokines (p < 0.05). In summary, we reported the functions of lnc-SPRR2G-2 and KHSRP in psoriasis. Our findings provide new insights for the further exploration of the pathogenesis and treatment of psoriasis.
Collapse
Affiliation(s)
- Yunyue Zhen
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xueqing Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Shan Huang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Ruijie Wang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Luan Yang
- Department of Dermatology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, Shandong, China
| | - Yingjian Huang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jianjun Yan
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Jiaoying Ju
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - He Wen
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
| | - Qing Sun
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
2
|
Gong W, Sun P, Li X, Wang X, Zhang X, Cui H, Yang J. Investigating the Molecular Mechanisms of Resveratrol in Treating Cardiometabolic Multimorbidity: A Network Pharmacology and Bioinformatics Approach with Molecular Docking Validation. Nutrients 2024; 16:2488. [PMID: 39125368 PMCID: PMC11314475 DOI: 10.3390/nu16152488] [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: 06/24/2024] [Revised: 07/14/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Resveratrol is a potent phytochemical known for its potential in treating cardiometabolic multimorbidity. However, its underlying mechanisms remain unclear. Our study systematically investigates the effects of resveratrol on cardiometabolic multimorbidity and elucidates its mechanisms using network pharmacology and molecular docking techniques. METHODS We screened cardiometabolic multimorbidity-related targets using the OMIM, GeneCards, and DisGeNET databases, and utilized the DSigDB drug characterization database to predict resveratrol's effects on cardiometabolic multimorbidity. Target identification for resveratrol was conducted using the TCMSP, SymMap, DrugBank, Swiss Target Prediction, CTD, and UniProt databases. SwissADME and ADMETlab 2.0 simulations were used to predict drug similarity and toxicity profiles of resveratrol. Protein-protein interaction (PPI) networks were constructed using Cytoscape 3.9.1 software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses were performed via the DAVID online platform, and target-pathway networks were established. Molecular docking validated interactions between core targets and resveratrol, followed by molecular dynamics simulations on the optimal core proteins identified through docking. Differential analysis using the GEO dataset validated resveratrol as a core target in cardiometabolic multimorbidity. RESULTS A total of 585 cardiometabolic multimorbidity target genes were identified, and the predicted results indicated that the phytochemical resveratrol could be a major therapeutic agent for cardiometabolic multimorbidity. SwissADME simulations showed that resveratrol has potential drug-like activity with minimal toxicity. Additionally, 6703 targets of resveratrol were screened. GO and KEGG analyses revealed that the main biological processes involved included positive regulation of cell proliferation, positive regulation of gene expression, and response to estradiol. Significant pathways related to MAPK and PI3K-Akt signaling pathways were also identified. Molecular docking and molecular dynamics simulations demonstrated strong interactions between resveratrol and core targets such as MAPK and EGFR. CONCLUSIONS This study predicts potential targets and pathways of resveratrol in treating cardiometabolic multimorbidity, offering a new research direction for understanding its molecular mechanisms. Additionally, it establishes a theoretical foundation for the clinical application of resveratrol.
Collapse
Affiliation(s)
- Wei Gong
- Public Health School, Ningxia Medical University, Yinchuan 750004, China; (W.G.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, China
- School of Medical Information and Engineering, Ningxia Medical University, Yinchuan 750004, China
| | - Peng Sun
- Public Health School, Ningxia Medical University, Yinchuan 750004, China; (W.G.)
- Science and Technology Center, Ningxia Medical University, Yinchuan 750001, China
- Ningxia Hui Autonomous Region Institute of Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiujing Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Xi Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Xinyu Zhang
- School of Medical Information and Engineering, Ningxia Medical University, Yinchuan 750004, China
| | - Huimin Cui
- Public Health School, Ningxia Medical University, Yinchuan 750004, China; (W.G.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, China
| | - Jianjun Yang
- Public Health School, Ningxia Medical University, Yinchuan 750004, China; (W.G.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, China
| |
Collapse
|
3
|
Hong L, Herjan T, Chen X, Zagore LL, Bulek K, Wang H, Yang CFJ, Licatalosi DD, Li X, Li X. Act1 drives chemoresistance via regulation of antioxidant RNA metabolism and redox homeostasis. J Exp Med 2024; 221:e20231442. [PMID: 38861022 PMCID: PMC11167376 DOI: 10.1084/jem.20231442] [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: 08/14/2023] [Revised: 02/20/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024] Open
Abstract
The IL-17 receptor adaptor molecule Act1, an RNA-binding protein, plays a critical role in IL-17-mediated cancer progression. Here, we report a novel mechanism of how IL-17/Act1 induces chemoresistance by modulating redox homeostasis through epitranscriptomic regulation of antioxidant RNA metabolism. Transcriptome-wide mapping of direct Act1-RNA interactions revealed that Act1 binds to the 5'UTR of antioxidant mRNAs and Wilms' tumor 1-associating protein (WTAP), a key regulator in m6A methyltransferase complex. Strikingly, Act1's binding sites are located in proximity to m6A modification sites, which allows Act1 to promote the recruitment of elF3G for cap-independent translation. Loss of Act1's RNA binding activity or Wtap knockdown abolished IL-17-induced m6A modification and translation of Wtap and antioxidant mRNAs, indicating a feedforward mechanism of the Act1-WTAP loop. We then developed antisense oligonucleotides (Wtap ASO) that specifically disrupt Act1's binding to Wtap mRNA, abolishing IL-17/Act1-WTAP-mediated antioxidant protein production during chemotherapy. Wtap ASO substantially increased the antitumor efficacy of cisplatin, demonstrating a potential therapeutic strategy for chemoresistance.
Collapse
Affiliation(s)
- Lingzi Hong
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tomasz Herjan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xing Chen
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Leah L. Zagore
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Katarzyna Bulek
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Han Wang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Donny D. Licatalosi
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Takeda Pharmaceutical Company, San Diego, CA, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiao Li
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Esfahani SK, Dehghani S, Hosseinzadeh H, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. An exosomal approach for oral delivery of resveratrol: Implications for inflammatory bowel disease treatment in rat model. Life Sci 2024; 346:122638. [PMID: 38614294 DOI: 10.1016/j.lfs.2024.122638] [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: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
AIMS Resveratrol (RSV) is a polyphenolic substance found in numerous natural products. Despite the wide range of therapeutic activities, including antioxidant and anti-inflammatory effects, the poor pharmacokinetic characteristics decrease the RSV bioavailability following oral administration. Milk-derived exosomes (MEXOs), as a class of natural nanocarriers, are promising candidates for oral drug delivery approaches. MAIN METHODS The current study developed RSV-loaded MEXOs to enhance the RSV oral bioavailability, introducing a suitable exosomal formulation for suppressing colon inflammation in acetic acid-induced rat models. KEY FINDINGS The results showed a remarkable encapsulation efficiency of 83.33 %. The in vitro release profile demonstrated a good retaining capability in acidic conditions (pH 1.2) and a considerable release in a simulated duodenal environment (pH 6.8). According to the permeability study, encapsulation of RSV improved its transportation across the Caco-2 monolayer. Moreover, the in vivo and histological analysis results proved that the RSV-MEXOs formulation successfully alleviates the inflammation in colitis rat models and effectively relieves the colitis. SIGNIFICANCE Our findings suggest that MEXOs should be of great attention as promising oral drug delivery vehicles for further clinical evaluations.
Collapse
Affiliation(s)
- Shaghayegh Kazemi Esfahani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Bolduan V, Palzer KA, Hieber C, Schunke J, Fichter M, Schneider P, Grabbe S, Pautz A, Bros M. The mRNA-Binding Protein KSRP Limits the Inflammatory Response of Macrophages. Int J Mol Sci 2024; 25:3884. [PMID: 38612694 PMCID: PMC11011855 DOI: 10.3390/ijms25073884] [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: 03/13/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
KH-type splicing regulatory protein (KSRP) is a single-stranded nucleic acid-binding protein with multiple functions. It is known to bind AU-rich motifs within the 3'-untranslated region of mRNA species, which in many cases encode dynamically regulated proteins like cytokines. In the present study, we investigated the role of KSRP for the immunophenotype of macrophages using bone marrow-derived macrophages (BMDM) from wild-type (WT) and KSRP-/- mice. RNA sequencing revealed that KSRP-/- BMDM displayed significantly higher mRNA expression levels of genes involved in inflammatory and immune responses, particularly type I interferon responses, following LPS stimulation. In line, time kinetics studies revealed increased levels of interferon-γ (IFN-γ), interleukin (IL)-1β and IL-6 mRNA in KSRP-/- macrophages after 6 h subsequent to LPS stimulation as compared to WT cultures. At the protein level, KSRP-/- BMDM displayed higher levels of these cytokines after overnight stimulation. Matching results were observed for primary peritoneal macrophages of KSRP-/- mice. These showed higher IL-6, tumor necrosis factor-α (TNF-α), C-X-C motif chemokine 1 (CXCL1) and CC-chemokine ligand 5 (CCL5) protein levels in response to LPS stimulation than the WT controls. As macrophages play a key role in sepsis, the in vivo relevance of KSRP deficiency for cytokine/chemokine production was analyzed in an acute inflammation model. In agreement with our in vitro findings, KSRP-deficient animals showed higher cytokine production upon LPS administration in comparison to WT mice. Taken together, these findings demonstrate that KSRP constitutes an important negative regulator of cytokine expression in macrophages.
Collapse
Affiliation(s)
- Vanessa Bolduan
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Kim-Alicia Palzer
- Department of Pharmacology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany (A.P.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany (A.P.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
6
|
Leavenworth JD, Yusuf N, Hassan Q. K-Homology Type Splicing Regulatory Protein: Mechanism of Action in Cancer and Immune Disorders. Crit Rev Eukaryot Gene Expr 2024; 34:75-87. [PMID: 37824394 PMCID: PMC11003564 DOI: 10.1615/critreveukaryotgeneexpr.2023048085] [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] [Indexed: 10/14/2023]
Abstract
K homology-type splicing regulatory protein (KSRP) is emerging as a key player in cancer biology, and immunology. As a single-strand nucleic acid binding protein it functions in both transcriptional and post-transcriptional regulation, while facilitating multiple stages of RNA metabolism to affect proliferation and control cell fate. However, it must interact with other proteins to determine the fate of its bound substrate. Here we provide an minireview of this important regulatory protein and describe its complex subcellular functions to affect RNA metabolism, stability, miRNA biogenesis and maturation, stress granule function, metastasis, and inflammatory processes.
Collapse
Affiliation(s)
- Jonathan D. Leavenworth
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nabiha Yusuf
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Quamarul Hassan
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
7
|
Cheng C, Cai Y, Liu X, Wu Y, Cheng Q, Wu Y, Wu Z. KHSRP modulated cell proliferation and cell cycle via regulating PPP2CA and p27 expression in Wilms tumor. Cell Signal 2022; 100:110447. [PMID: 36029941 DOI: 10.1016/j.cellsig.2022.110447] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/15/2022]
Abstract
Wilms tumor (WT) is the most common renal malignancy in children, and the survival rate of high-risk WT patients was still low despite multimodality therapy. KHSRP, an RNA-binding protein, has been proved to be relative to tumor progression in different kinds of malignancies, but the function of KHSRP in WT remained unclear. Here, our study aimed to explore and clarify the function of KHSRP in WT cells and its molecular mechanism. Thus, our results showed that KHSRP was highly expressed in WT tumor tissues compared to normal kidney tissues and correlated with poor prognosis in WT patients. Downregulation of KHSRP using siRNAs in WT cell line SK-NEP-1 and Wit49 resulted in inhibition of cell proliferation and cell cycle arrest via stabilizing and upregulating p27 protein. Furthermore, mechanistic analyses revealed that KHSRP bound to 3'UTR of PPP2CA mRNA and modulating its mRNA stability, resulting in regulation of the phosphorylation level and protein stability of p27 in WT cell lines. In conclusion, our results demonstrated that KHSRP played an important role in WT and modulated cell proliferation and cell cycle via regulating the expression of PPP2CA and p27.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China
| | - Yuanxia Cai
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China
| | - Xiaowei Liu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China
| | - Yangkun Wu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China
| | - Qianqian Cheng
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China
| | - Yeming Wu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China; Department of Pediatric Surgery, Hangzhou Children's Hospital, Hangzhou, Hangzhou 310010, China.
| | - Zhixiang Wu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200092 Shanghai, China; Division of Pediatric Oncology, Shanghai Institute of Pediatric Research, 200092 Shanghai, China; Department of Pediatric Surgery, Children's Hospital of Soochow University, 215003 Suzhou, China.
| |
Collapse
|
8
|
Nutrigenomics: An inimitable interaction amid genomics, nutrition and health. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2022.103196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
9
|
Seyyed Shoura SM, Naghsh N, Moslemi E, Kavyani Z, Moridpour AH, Musazadeh V, Dehghan P. Can resveratrol supplementation affect biomarkers of inflammation and oxidative stress? An umbrella meta-analysis. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
|
10
|
Wan JT, Qiu XS, Fu ZH, Huang YC, Min SX. Tumor necrosis factor-α inhibition restores matrix formation by human adipose-derived stem cells in the late stage of chondrogenic differentiation. World J Stem Cells 2022; 14:798-814. [PMID: 36483847 PMCID: PMC9724386 DOI: 10.4252/wjsc.v14.i11.798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/05/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering is a promising strategy for treating cartilage damage. Matrix formation by adipose-derived stem cells (ADSCs), which are one type of seed cell used for cartilage tissue engineering, decreases in the late stage of induced chondrogenic differentiation in vitro, which seriously limits research on ADSCs and their application.
AIM To improve the chondrogenic differentiation efficiency of ADSCs in vitro, and optimize the existing chondrogenic induction protocol.
METHODS Tumor necrosis factor-alpha (TNF-α) inhibitor was added to chondrogenic culture medium, and then Western blotting, enzyme linked immunosorbent assay, immunofluorescence and toluidine blue staining were used to detect the cartilage matrix secretion and the expression of key proteins of nuclear factor kappa-B (NF-κB) signaling pathway.
RESULTS In this study, we found that the levels of TNF-α and matrix metalloproteinase 3 were increased during the chondrogenic differentiation of ADSCs. TNF-α then bound to its receptor and activated the NF-κB pathway, leading to a decrease in cartilage matrix synthesis and secretion. Blocking TNF-α with its inhibitors etanercept (1 μg/mL) or infliximab (10 μg/mL) significantly restored matrix formation.
CONCLUSION Therefore, this study developed a combination of ADSC therapy and targeted anti-inflammatory drugs to optimize the chondrogenesis of ADSCs, and this approach could be very beneficial for translating ADSC-based approaches to treat cartilage damage.
Collapse
Affiliation(s)
- Jiang-Tao Wan
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- Institute of Orthopedics, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong Province, China
| | - Xian-Shuai Qiu
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Zhuo-Hang Fu
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- Institute of Orthopedics, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong Province, China
| | - Yong-Can Huang
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- Institute of Orthopedics, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong Province, China
| | - Shao-Xiong Min
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| |
Collapse
|
11
|
Targeting the "undruggable": RNA-binding proteins in the spotlight in cancer therapy. Semin Cancer Biol 2022; 86:69-83. [PMID: 35772609 DOI: 10.1016/j.semcancer.2022.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/19/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023]
Abstract
Tumors refractory to conventional therapy belong to specific subpopulations of cancer cells, which have acquired a higher number of mutations/epigenetic changes than the majority of cancer cells. This property provides them the ability to become resistant to therapy. Aberrant expression of certain RNA-binding proteins (RBPs) can regulate the sensitivity of tumor cells to chemotherapeutic drugs by binding to specific regions present in the 3´-UTR of certain mRNAs to promote or repress mRNA translation or by interacting with other proteins (including RBPs) and non-coding RNAs that are part of ribonucleoprotein complexes. In particular, an increasing interest in the RBPs involved in chemoresistance has recently emerged. In this review, we discuss how RBPs are not only affected by chemotherapeutic treatments, but also play an active role in therapeutic responses via the direct modulation of crucial cancer-related proteins. A special focus is being placed on the development of therapeutic strategies targeting these RBPs.
Collapse
|
12
|
Singh S, Shaikh IA, More SS, Mahnashi MH, Almohaimeed HM, El-Sherbiny M, Ghoneim MM, Umar A, Soni HK, Agrawal H, Mannasaheb BA, Khan AA, Muddapur UM, Iqubal SMS. Blockage of KHSRP-NLRP3 by MCC950 Can Reverse the Effect of Manganese-Induced Neuroinflammation in N2a Cells and Rat Brain. Int J Mol Sci 2022; 23:13224. [PMID: 36362011 PMCID: PMC9658363 DOI: 10.3390/ijms232113224] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 01/09/2024] Open
Abstract
Manganese neurotoxicity has been reported to cause a neurodegenerative disease known as parkinsonism. Previous reports have shown that the expression of the KH-type splicing regulatory protein (KHSRP), a nucleic acid-binding protein, and NLRP3 is increased upon Mn exposure. However, the relation between these two during Mn toxicity has not been fully deduced. The mouse neuroblastoma (N2a) and SD rats are treated with LPS and MnCl2 to evaluate the expression of KHSRP and NLRP3. Further, the effect of the NLRP3 inhibitor MCC950 is checked on the expression of NLRP3, KHSRP and pro-inflammatory markers (TNFα, IL-18 and IL-1β) as well as the caspase-1 enzyme. Our results demonstrated an increment in NLRP3 and KHSRP expression post-MnCl2 exposure in N2a cells and rat brain, while on the other hand with LPS exposure only NLRP3 expression levels were elevated and KHSRP was found to be unaffected. An increased expression of KHSRP, NLRP3, pro-inflammatory markers and the caspase-1 enzyme was observed to be inhibited with MCC950 treatment in MnCl2-exposed cells and rats. Manganese exposure induces NLRP3 and KHSRP expression to induce neuroinflammation, suggesting a correlation between both which functions in toxicity-related pathways. Furthermore, MCC950 treatment reversed the role of KHSRP from anti-inflammatory to pro-inflammatory.
Collapse
Affiliation(s)
- Sharad Singh
- School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore 560111, Karnataka, India
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, P.O. Box 1988, Najran 66462, Saudi Arabia
| | - Sunil S. More
- School of Basic and Applied Sciences, Dayananda Sagar University, Bangalore 560111, Karnataka, India
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, P.O. Box 1988, Najran 66462, Saudi Arabia
| | - Hailah M. Almohaimeed
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohammed M. Ghoneim
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Ahmad Umar
- Department of Chemistry, Faculty of Science and Arts, Najran University, P.O. Box 1988, Najran 11001, Saudi Arabia
- Promising Centre for Sensors and Electronic Devices (PCSED), Najran University, P.O. Box 1988, Najran 11001, Saudi Arabia
| | - Harshit Kumar Soni
- Department of Zoology, Government Science College, Pandhurna 480334, Madhya Pradesh, India
| | - Himanshu Agrawal
- Jubilant Biosys Limited (Discovery Biology), Bangalore 560022, Karnataka, India
| | - Basheer Ahmed Mannasaheb
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Aejaz Abdullatif Khan
- Department of General Science, Ibn Sina National College for Medical Studies, P.O. Box 31906, Jeddah 21418, Saudi Arabia
| | - Uday M. Muddapur
- Department of Biotechnology, KLE Technological University, BVB Campus, Hubballi 580031, Karnataka, India
| | - S. M. Shakeel Iqubal
- Department of General Science, Ibn Sina National College for Medical Studies, P.O. Box 31906, Jeddah 21418, Saudi Arabia
| |
Collapse
|
13
|
Palzer KA, Bolduan V, Käfer R, Kleinert H, Bros M, Pautz A. The Role of KH-Type Splicing Regulatory Protein (KSRP) for Immune Functions and Tumorigenesis. Cells 2022; 11:cells11091482. [PMID: 35563788 PMCID: PMC9104899 DOI: 10.3390/cells11091482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Post-transcriptional control of gene expression is one important mechanism that enables stringent and rapid modulation of cytokine, chemokines or growth factors expression, all relevant for immune or tumor cell function and communication. The RNA-binding protein KH-type splicing regulatory protein (KSRP) controls the mRNA stability of according genes by initiation of mRNA decay and inhibition of translation, and by enhancing the maturation of microRNAs. Therefore, KSRP plays a pivotal role in immune cell function and tumor progression. In this review, we summarize the current knowledge about KSRP with regard to the regulation of immunologically relevant targets, and the functional role of KSRP on immune responses and tumorigenesis. KSRP is involved in the control of myeloid hematopoiesis. Further, KSRP-mediated mRNA decay of pro-inflammatory factors is necessary to keep immune homeostasis. In case of infection, functional impairment of KSRP is important for the induction of robust immune responses. In this regard, KSRP seems to primarily dampen T helper cell 2 immune responses. In cancer, KSRP has often been associated with tumor growth and metastasis. In summary, aside of initiation of mRNA decay, the KSRP-mediated regulation of microRNA maturation seems to be especially important for its diverse biological functions, which warrants further in-depth examination.
Collapse
Affiliation(s)
- Kim-Alicia Palzer
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.-A.P.); (R.K.); (H.K.)
| | - Vanessa Bolduan
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (V.B.); (M.B.)
| | - Rudolf Käfer
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.-A.P.); (R.K.); (H.K.)
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.-A.P.); (R.K.); (H.K.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (V.B.); (M.B.)
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.-A.P.); (R.K.); (H.K.)
- Correspondence: ; Tel.: +49-6131-179276; Fax: +49-6131-179042
| |
Collapse
|
14
|
Saurin S, Meineck M, Erkel G, Opatz T, Weinmann-Menke J, Pautz A. Drug Candidates for Autoimmune Diseases. Pharmaceuticals (Basel) 2022; 15:503. [PMID: 35631330 PMCID: PMC9143092 DOI: 10.3390/ph15050503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 12/10/2022] Open
Abstract
Most of the immunosuppressive drugs used in the clinic to prevent organ rejection or to treat autoimmune disorders were originally isolated from fungi or bacteria. Therefore, in addition to plants, these are valuable sources for identification of new potent drugs. Many side effects of established drugs limit their usage and make the identification of new immunosuppressants necessary. In this review, we present a comprehensive overview of natural products with potent anti-inflammatory activities that have been tested successfully in different models of chronic inflammatory autoimmune diseases. Some of these candidates already have passed first clinical trials. The anti-inflammatory potency of these natural products was often comparable to those of established drugs, and they could be used at least in addition to standard therapy to reduce their dose to minimize unwanted side effects. A frequent mode of action is the inhibition of classical inflammatory signaling pathways, such as NF-κB, in combination with downregulation of oxidative stress. A drawback for the therapeutic use of those natural products is their moderate bioavailability, which can be optimized by chemical modifications and, in addition, further safety studies are necessary. Altogether, very interesting candidate compounds exist which have the potential to serve as starting points for the development of new immunosuppressive drugs.
Collapse
Affiliation(s)
- Sabrina Saurin
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.S.); (M.M.)
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Myriam Meineck
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.S.); (M.M.)
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Gerhard Erkel
- Department of Molecular Biotechnology and Systems Biology, Technical University, 67663 Kaiserslautern, Germany;
| | - Till Opatz
- Department of Chemistry, Johannes Gutenberg University, 55099 Mainz, Germany;
| | - Julia Weinmann-Menke
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.S.); (M.M.)
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
15
|
De Pieri A, Ocorr K, Jerreld K, Lamoca M, Hitzl W, Wuertz-Kozak K. Resveratrol Microencapsulation into Electrosprayed Polymeric Carriers for the Treatment of Chronic, Non-Healing Wounds. Pharmaceutics 2022; 14:pharmaceutics14040853. [PMID: 35456686 PMCID: PMC9031663 DOI: 10.3390/pharmaceutics14040853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic, non-healing wounds represent a challenging socio-economic burden, demanding innovative approaches for successful wound management. Resveratrol (RSV) represents a promising therapeutic candidate, but its therapeutic efficacy and clinical applicability have been hampered by its rapid degradation and/or depletion. Herein, RSV was encapsulated into poly(ε-caprolactone) (PCL) microparticles by electrospraying with the aim to prolong and preserve RSV’s release/activity, without affecting its therapeutic properties. Electrospraying led to the fabrication of spherical (2 to 10 μm in size), negatively charged (<−1 mV), and quasi-monodisperse (PDI < 0.3) microparticles, with 60% RSV release after 28 days. Microencapsulation of RSV into PCL prevented its photochemical degradation and preserved its antioxidant properties over 72 h. The RSV-PCL microparticles did not exhibit any cytotoxicity on human dermal fibroblasts. RSV released from the microparticles was biologically functional and induced a significant increase in collagen type I deposition. Furthermore, the produced RSV-PCL microparticles reduced the expression of inflammatory (IL-6, IL-8, COX-2) and proteolytic (MMP-2, MMP-9) mediators. Collectively, our data clearly illustrate the potential of electrosprayed polymeric carriers for the sustained delivery of RSV to treat chronic wounds.
Collapse
Affiliation(s)
- Andrea De Pieri
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY 14623, USA; (A.D.P.); (K.O.); (K.J.); (M.L.)
| | - Keegan Ocorr
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY 14623, USA; (A.D.P.); (K.O.); (K.J.); (M.L.)
| | - Kyle Jerreld
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY 14623, USA; (A.D.P.); (K.O.); (K.J.); (M.L.)
| | - Mikkael Lamoca
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY 14623, USA; (A.D.P.); (K.O.); (K.J.); (M.L.)
| | - Wolfgang Hitzl
- Research and Innovation Management (RIM), Biostatistics and Publication of Clinical Trial Studies, Paracelsus Medical University, 5020 Salzburg, Austria;
- Department of Ophthalmology and Optometry, Paracelsus Medical University, 5020 Salzburg, Austria
- Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY 14623, USA; (A.D.P.); (K.O.); (K.J.); (M.L.)
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), 81547 Munich, Germany
- Correspondence: ; Tel.: +1-(585)-475-7355
| |
Collapse
|
16
|
Gather F, Ihrig-Biedert I, Kohlhas P, Krutenko T, Peitz M, Brüstle O, Pautz A, Kleinert H. A specific, non-immune system-related isoform of the human inducible nitric oxide synthase is expressed during differentiation of human stem cells into various cell types. Cell Commun Signal 2022; 20:47. [PMID: 35392923 PMCID: PMC8991583 DOI: 10.1186/s12964-022-00855-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND NOS2 expression is mostly found in bacteria-exposed or cytokine-treated tissues and is mostly connected to innate immune reactions. There are three isoforms of NOS2 (NOS2-1 to -3). In RNA-seq data sets, analyzing inflammatory gene expression, only expression of the NOS2-1 mRNA isoform is detected. However, the expression of NOS2 in differentiating human pluripotent stems (hPSCs) has not been analyzed yet. METHODS Public available RNA-seq databases were screened for data of hPSCs during differentiation to different target cells. An isoform specific algorithm was used to analyze NOS2 mRNA isoform expression. In addition, we differentiated four different human iPSC cell lines toward cortical neurons and analyzed NOS2 mRNA expression by qRT-PCR and 5'-RACE. The functionality of the NOS2-2 protein was analyzed by transient transfection of expression clones in human DLD1 cells and nitrate measurement in the supernatant of these cells. RESULTS In RNA-seq databases we detected a transient expression of the NOS2 mRNA during the differentiation of hPSCs to cardiomyocytes, chondrocytes, mesenchymal stromal cells, neurons, syncytiotrophoblast cells, and trophoblasts. NOS2 mRNA isoform specific analyses showed, that the transiently expressed NOS2 mRNA in differentiating hPSC (NOS2-2; "diff-iNOS") differ remarkably from the already described NOS2 transcript found in colon or induced islets (NOS2-1; "immuno-iNOS"). Also, analysis of the NOS2 mRNA- and protein expression during the differentiation of four different hiPSC lines towards cortical neurons showed a transient expression of the NOS2 mRNA and NOS2 protein on day 18 of the differentiation course. 5'-RACE experiments and isoform specific qRT-PCR analyses revealed that only the NOS2-2 mRNA isoform was expressed in these experiments. To analyze the functionality of the NOS2-2 protein, we transfected human DLD-1 cells with tetracycline inducible expression clones encoding the NOS2-1- or -2 coding sequence. After induction of the NOS2-1 or -2 mRNA expression by tetracycline a similar nitrate production was measured proofing the functionality of the NOS2-2 protein isoform. CONCLUSIONS Our data show that a differentiation specific NOS2 isoform (NOS2-2) is transiently expressed during differentiation of hPSC. Video Abstract.
Collapse
Affiliation(s)
- Fabian Gather
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Department of Molecular Embryology, Institute for Anatomy and Cell Biology, Freiburg, Germany
| | - Irmgard Ihrig-Biedert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Paul Kohlhas
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Tamara Krutenko
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Michael Peitz
- Cell Programming Core Facility, Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.,Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
17
|
Barth E, Srivastava A, Wengerodt D, Stojiljkovic M, Axer H, Witte OW, Kretz A, Marz M. Age-dependent expression changes of circadian system-related genes reveal a potentially conserved link to aging. Aging (Albany NY) 2021; 13:25694-25716. [PMID: 34923482 PMCID: PMC8751596 DOI: 10.18632/aging.203788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022]
Abstract
The circadian clock system influences the biology of life by establishing circadian rhythms in organisms, tissues, and cells, thus regulating essential biological processes based on the day/night cycle. Circadian rhythms change over a lifetime due to maturation and aging, and disturbances in the control of the circadian system are associated with several age-related pathologies. However, the impact of chronobiology and the circadian system on healthy organ and tissue aging remains largely unknown. Whether aging-related changes of the circadian system’s regulation follow a conserved pattern across different species and tissues, hence representing a common driving force of aging, is unclear. Based on a cross-sectional transcriptome analysis covering 329 RNA-Seq libraries, we provide indications that the circadian system is subjected to aging-related gene alterations shared between evolutionarily distinct species, such as Homo sapiens, Mus musculus, Danio rerio, and Nothobranchius furzeri. We discovered differentially expressed genes by comparing tissue-specific transcriptional profiles of mature, aged, and old-age individuals and report on six genes (per2, dec2, cirp, klf10, nfil3, and dbp) of the circadian system, which show conserved aging-related expression patterns in four organs of the species examined. Our results illustrate how the circadian system and aging might influence each other in various tissues over a long lifespan and conceptually complement previous studies tracking short-term diurnal and nocturnal gene expression oscillations.
Collapse
Affiliation(s)
- Emanuel Barth
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany
| | - Akash Srivastava
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany.,FLI Leibniz Institute for Age Research, Jena, Germany.,Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Diane Wengerodt
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Milan Stojiljkovic
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Hubertus Axer
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Alexandra Kretz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Manja Marz
- Bioinformatics/High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich Schiller University Jena, Jena, Germany.,FLI Leibniz Institute for Age Research, Jena, Germany.,German Center for Integrative Biodiversity Research (iDiv), Halle-Jena-Leipzig, Germany.,European Virus Bioinformatics Center (EVBC), Jena, Germany
| |
Collapse
|
18
|
Schmidtke L, Meineck M, Saurin S, Otten S, Gather F, Schrick K, Käfer R, Roth W, Kleinert H, Weinmann-Menke J, Pautz A. Knockout of the KH-Type Splicing Regulatory Protein Drives Glomerulonephritis in MRL-Fas lpr Mice. Cells 2021; 10:3167. [PMID: 34831390 PMCID: PMC8624031 DOI: 10.3390/cells10113167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
KH-type splicing regulatory protein (KSRP) is an RNA-binding protein that promotes mRNA decay and thereby negatively regulates cytokine expression at the post-transcriptional level. Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated cytokine expression causing multiple organ manifestations; MRL-Faslpr mice are an established mouse model to study lupus disease pathogenesis. To investigate the impact of KSRP on lupus disease progression, we generated KSRP-deficient MRL-Faslpr mice (MRL-Faslpr/KSRP-/- mice). In line with the predicted role of KSRP as a negative regulator of cytokine expression, lupus nephritis was augmented in MRL-Faslpr/KSRP-/- mice. Increased infiltration of immune cells, especially of IFN-γ producing T cells and macrophages, driven by enhanced expression of T cell-attracting chemokines and adhesion molecules, seems to be responsible for worsened kidney morphology. Reduced expression of the anti-inflammatory interleukin-1 receptor antagonist may be another reason for severe inflammation. The increase of FoxP3+ T cells detected in the kidney seems unable to dampen the massive kidney inflammation. Interestingly, lymphadenopathy was reduced in MRL-Faslpr/KSRP-/- mice. Altogether, KSRP appears to have a complex role in immune regulation; however, it is clearly able to ameliorate lupus nephritis.
Collapse
Affiliation(s)
- Lisa Schmidtke
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Myriam Meineck
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Sabrina Saurin
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Svenja Otten
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Fabian Gather
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Katharina Schrick
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Rudolf Käfer
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Julia Weinmann-Menke
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| |
Collapse
|
19
|
Omraninava M, Razi B, Aslani S, Imani D, Jamialahmadi T, Sahebkar A. Effect of resveratrol on inflammatory cytokines: A meta-analysis of randomized controlled trials. Eur J Pharmacol 2021; 908:174380. [PMID: 34303665 DOI: 10.1016/j.ejphar.2021.174380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/25/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022]
Abstract
The aim of the current study was to perform a meta-analysis of randomized clinical trials regarding the effect of resveratrol in decreasing the levels of inflammatory cytokines, including interleukin (IL)-1, IL-6, IL-8, and tumor necrosis factor (TNF)-α in a combination of inflammatory diseases. Literature search was carried out in Scopus, ISI web of science, Medline, and Cochrane Library databases by up to September 2020. The pooled effect size was determined through measuring the weighted mean differences (WMD) and their corresponding 95% confidence intervals (CI) for the difference between the resveratrol-receiving and control groups. Finally, 33 publications, including 3 studies on IL-1, 26 studies on IL-6, 4 studies on IL-8, and 21 studies on TNF-α met our final inclusion criteria and included in the quantitative analysis. Analysis in the overall population showed a significant effect of resveratrol consumption in reducing serum TNF-α levels (WMD = -0.66 pg/ml, 95% CI = -1.05 to -0.27, P = 0.001). A significant reduction of IL-6 concentration was observed only in the patients receiving ≥500 mg/day dose of resveratrol (WMD = -1.89 pg/ml, 95% CI = -3.73 to -0.05, P = 0.04) with inter-study heterogeneity (I2 = 94.4%, P < 0.001). Nonetheless, no significant alteration was observed in IL-1 (WMD = -0.14 pg/ml, 95% CI = -0.31 to 0.03, P = 0.10) and IL-8 (WMD = 0.18 pg/ml, 95% CI = -1.04 to 1.40, P = 0.73) levels following resveratrol consumption. Based on the present findings, resveratrol is able to decrease TNF-α and IL-6 (in ≥500 mg/day dose) levels but not IL-1 and IL-8 levels.
Collapse
Affiliation(s)
- Melodi Omraninava
- Department of Infectious Disease, Faculty of Medical Sciences, Sari Branch, Islamic Azad University, Sari, Iran
| | - Bahman Razi
- Department of Hematology and Blood Banking, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Danyal Imani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Abstract
Posttranscriptional control of mRNA regulates various biological processes, including inflammatory and immune responses. RNA-binding proteins (RBPs) bind cis-regulatory elements in the 3' untranslated regions (UTRs) of mRNA and regulate mRNA turnover and translation. In particular, eight RBPs (TTP, AUF1, KSRP, TIA-1/TIAR, Roquin, Regnase, HuR, and Arid5a) have been extensively studied and are key posttranscriptional regulators of inflammation and immune responses. These RBPs sometimes collaboratively or competitively bind the same target mRNA to enhance or dampen regulatory activities. These RBPs can also bind their own 3' UTRs to negatively or positively regulate their expression. Both upstream signaling pathways and microRNA regulation shape the interactions between RBPs and target RNA. Dysregulation of RBPs results in chronic inflammation and autoimmunity. Here, we summarize the functional roles of these eight RBPs in immunity and their associated diseases.
Collapse
Affiliation(s)
- Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0874, Japan.,Department of Host Defense, Division of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0874, Japan;
| | - Kazuhiko Maeda
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0874, Japan.,Department of Host Defense, Division of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0874, Japan;
| |
Collapse
|
21
|
Begum D, Merchant N, Nagaraju GP. Role of selected phytochemicals on gynecological cancers. A THERANOSTIC AND PRECISION MEDICINE APPROACH FOR FEMALE-SPECIFIC CANCERS 2021:1-30. [DOI: 10.1016/b978-0-12-822009-2.00001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Effects of Resveratrol on Thymic Stromal Lymphopoietin Expression in Mast Cells. ACTA ACUST UNITED AC 2020; 57:medicina57010021. [PMID: 33379407 PMCID: PMC7824250 DOI: 10.3390/medicina57010021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/20/2022]
Abstract
Background and objectives: Cytokine thymic stromal lymphopoietin (TSLP) plays a pivotal role in the pathogenesis of atopic diseases such as atopic dermatitis, allergic rhinitis, and asthma. Resveratrol (RSV) exerts various pharmacological effects such as antioxidant, anti-inflammatory, neuroprotective, and anticancer. Although, it has been verified the beneficial effects of RSV on various subjects, the effect of RSV on thymic stromal lymphopoietin (TSLP) regulation has not been elucidated. Materials and Methods: Here, we examined how RSV regulates TSLP in HMC-1 cells. Enzyme-linked immunosorbent assay, real-time polymerase chain reaction, Western blotting, and calcium assay were performed to evaluate the effect of RSV. Results: TSLP production and mRNA expression were reduced by RSV. RSV down-regulated nuclear factor-κB activation, IκBα phosphorylation as well as activation of receptor-interacting protein2 and caspase-1 in HMC-1 cells. In addition, RSV treatment decreased the up-regulation of intracellular calcium in HMC-1 cells. Conclusions: These results suggest that RSV might be useful for the treatment of atopic diseases through blocking of TSLP.
Collapse
|
23
|
Wang C, Xu W, Chao Y, Liang M, Zhang F, Huang K. E3 Ligase FBXW2 Is a New Therapeutic Target in Obesity and Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001800. [PMID: 33101872 PMCID: PMC7578860 DOI: 10.1002/advs.202001800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/02/2020] [Indexed: 05/05/2023]
Abstract
Chronic low-grade inflammation orchestrated by macrophages plays a critical role in metabolic chronic diseases, like obesity and atherosclerosis. However, the underlying mechanism remains to be elucidated. Here, the E3 ubiquitin ligase F-box/WD Repeat-Containing Protein 2 (FBXW2), the substrate-binding subunit of E3 ubiquitin ligase SCF (a complex of FBXW2, SKP1, and cullin-1), as an inflammatory mediator in macrophages, is identified. Myeloid-specific FBXW2 gene deficiency improves both obesity-associated with insulin resistance and atherosclerosis in murine models. The beneficial effects by FBXW2 knockout are accompanied by decreased proinflammatory responses and macrophage infiltration in the microenvironment. Mechanistically, it is identified that KH-type splicing regulatory protein (KSRP) is a new bona fide ubiquitin substrate of SCFFBXW2. Inhibition of KSRP prevents FBXW2-deficient macrophages from exerting a protective effect on inflammatory reactions, insulin resistance and plaque formation. Furthermore, it is demonstrated that the C-terminus (P3) of FBXW2 competitively ablates the function of FBXW2 in KSRP degradation and serves as an effective inhibitor of obesity and atherogenesis progression. Thus, the data strongly suggest that SCFFBXW2 is an important mediator in the context of metabolic diseases. The development of FBXW2 (P3)-mimicking inhibitors and small-molecular drugs specifically abrogating KSRP ubiquitination-dependent inflammatory responses are viable approaches for obesity and atherosclerosis treatment.
Collapse
Affiliation(s)
- Cheng Wang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Department of RheumatologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Wenjing Xu
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yuelin Chao
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjing210029China
| | - Minglu Liang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Fengxiao Zhang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Kai Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
24
|
Nuclear P38: Roles in Physiological and Pathological Processes and Regulation of Nuclear Translocation. Int J Mol Sci 2020; 21:ijms21176102. [PMID: 32847129 PMCID: PMC7504396 DOI: 10.3390/ijms21176102] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
The p38 mitogen-activated protein kinase (p38MAPK, termed here p38) cascade is a central signaling pathway that transmits stress and other signals to various intracellular targets in the cytoplasm and nucleus. More than 150 substrates of p38α/β have been identified, and this number is likely to increase. The phosphorylation of these substrates initiates or regulates a large number of cellular processes including transcription, translation, RNA processing and cell cycle progression, as well as degradation and the nuclear translocation of various proteins. Being such a central signaling cascade, its dysregulation is associated with many pathologies, particularly inflammation and cancer. One of the hallmarks of p38α/β signaling is its stimulated nuclear translocation, which occurs shortly after extracellular stimulation. Although p38α/β do not contain nuclear localization or nuclear export signals, they rapidly and robustly translocate to the nucleus, and they are exported back to the cytoplasm within minutes to hours. Here, we describe the physiological and pathological roles of p38α/β phosphorylation, concentrating mainly on the ill-reviewed regulation of p38α/β substrate degradation and nuclear translocation. In addition, we provide information on the p38α/β ’s substrates, concentrating mainly on the nuclear targets and their role in p38α/β functions. Finally, we also provide information on the mechanisms of nuclear p38α/β translocation and its use as a therapeutic target for p38α/β-dependent diseases.
Collapse
|
25
|
Lourou N, Gavriilidis M, Kontoyiannis DL. Lessons from studying the AU-rich elements in chronic inflammation and autoimmunity. J Autoimmun 2019; 104:102334. [PMID: 31604649 DOI: 10.1016/j.jaut.2019.102334] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
AU-rich elements (AREs) comprise one of the most widely studied families of regulatory RNA structures met in RNAs engaged in complex immunological reactions. A multitude of genetic, molecular, holistic and functional studies have been utilized for the analyses of the AREs and their interactions to proteins that bind to them. Data stemming from these studies brought forth a world of RNA-related check-points against infection, chronic inflammation, tumor associated immunity, and autoimmunity; and the interest to capitalize the interactions of AREs for clinical management and therapy. They also provided lessons on the cellular capabilities of post-transcriptional control. Originally thought as transcript-restricted regulators of turnover and translation, ARE-binding proteins do in fact harbor great versatility and interactivity across nuclear and cytoplasmic compartments; and act as functional coordinators of immune-cellular programs. Harnessing these deterministic functions requires extensive knowledge of their synergies or antagonisms at a cell-specific level; but holds great promise since it can provide the efficacy of combinatorial therapies with single agents.
Collapse
Affiliation(s)
- Niki Lourou
- School of Biology, Department of Development, Genetics and Molecular Biology, Aristotle University of Thessaloniki, Greece
| | - Maxim Gavriilidis
- School of Biology, Department of Development, Genetics and Molecular Biology, Aristotle University of Thessaloniki, Greece; Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Greece
| | - Dimitris L Kontoyiannis
- School of Biology, Department of Development, Genetics and Molecular Biology, Aristotle University of Thessaloniki, Greece; Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Greece.
| |
Collapse
|
26
|
The RNA-Binding Protein KSRP Modulates Cytokine Expression of CD4 + T Cells. J Immunol Res 2019; 2019:4726532. [PMID: 31511826 PMCID: PMC6714327 DOI: 10.1155/2019/4726532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/13/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023] Open
Abstract
The KH-type splicing regulatory protein (KSRP) is a RNA-binding protein, which regulates the stability of many mRNAs encoding immune-relevant proteins. As KSRP regulates innate immune responses, for instance by the modulation of type I interferon mRNA stability, we were interested whether knockdown of the protein (KSRP−/−) interferes with T cell activation and polarization. Polyclonally stimulated KSRP−/− CD4+ T cells proliferated at a higher extent and higher frequency and expressed the activation marker CD25 more than wild-type T cells. In supernatants of stimulated KSRP−/− CD4+ T cells, levels of IL-5, IL-9, IL-10, and IL-13 were observed to be increased compared to those of the control group. KSRP−/− CD8+ T cells showed no altered proliferative capacity upon polyclonal stimulation, but supernatants contained lower levels of interferon-γ. Similar changes in the cytokine expression patterns were also detected in T cells derived from KSRP−/− mice undergoing arthritis induction indicative of a pathophysiological role of KSRP-dependent T cell polarization. We demonstrated the direct binding of KSRP to the 3′ untranslated region of IL-13, IL-10, and IFN-γ mRNA in in vitro experiments. Moreover, since IL-4 mRNA decay was reduced in KSRP−/− CD4+ T cells, we identify KSRP as a negative regulator of IL-4 expression. These data indicate that overexpression of IL-4, which constitutes the primary inducer of Th2 polarization, may cause the Th2 bias of polyclonally stimulated KSRP−/− CD4+ T cells. This is the first report demonstrating that KSRP is involved in the regulation of T cell responses. We present strong evidence that T cells derived from KSRP−/− mice favor Th2-driven immune responses.
Collapse
|
27
|
Gather F, Schmitz K, Koch K, Vogt LM, Pautz A, Kleinert H. Regulation of human inducible nitric oxide synthase expression by an upstream open reading frame. Nitric Oxide 2019; 88:50-60. [PMID: 31004763 DOI: 10.1016/j.niox.2019.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/04/2019] [Accepted: 04/15/2019] [Indexed: 01/05/2023]
Abstract
The human inducible nitric oxide synthase (iNOS) gene contains an upstream open reading frame (uORF) in its 5'-untranslated region (5'-UTR) implying a translational regulation of iNOS expression. Transfection experiments in human DLD-1 cells revealed that the uORF although translatable seems not to inhibit the translation start at the bona fide ATG. Our data clearly show that human iNOS translation is cap-dependent and that the 5'-UTR of the iNOS mRNA contains no internal ribosome entry site. Translation of the bona fide coding sequence is most likely mediated by a leaky scanning mechanism. The 5'-UTR is encoded by exon 1 and exon 2 of the iNOS gene with the uORF stop codon located in front of the first intron indicating an involvement of the nonsense mediated RNA decay (NMD) in iNOS regulation. SiRNA-mediated down-regulation of Upf1 resulted in enhanced endogenous cytokine iNOS expression in human DLD-1 cells. Transfection of constructs containing iNOS exon 1, intron 1 and exon 2 in front of a luciferase gene showed a clear effect of the mutation of the uORF-ATG on luciferase reportergene expression. Our data indicate that the uORF in the 5'-UTR sequence of human iNOS gene reduces its expression via the NMD mechanism.
Collapse
Affiliation(s)
- Fabian Gather
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany
| | - Katja Schmitz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany
| | - Kathrin Koch
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany
| | - Lea-Marie Vogt
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany.
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher Str. 67, 55101, Mainz, Germany.
| |
Collapse
|
28
|
Hartung NM, Fischer J, Ostermann AI, Willenberg I, Rund KM, Schebb NH, Garscha U. Impact of food polyphenols on oxylipin biosynthesis in human neutrophils. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1536-1544. [PMID: 31125704 DOI: 10.1016/j.bbalip.2019.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/11/2019] [Accepted: 05/10/2019] [Indexed: 12/29/2022]
Abstract
The intake of food polyphenols is associated with beneficial impacts on health. Besides anti-oxidative effects, anti-inflammatory properties have been suggested as molecular modes of action, which may result from modulations of the arachidonic acid (AA) cascade. Here, we investigated the effects of a library of food polyphenols on 5-lipoxygenase (5-LOX) activity in a cell-free assay, and in human neutrophils. Resveratrol, its dimer (ε-viniferin), and its imine analogue (IRA) potently blocked the 5-LOX-mediated LT formation in neutrophils with IC50 values in low μM-range. Among the tested flavonoids only the isoflavone genistein showed potent 5-LOX inhibition in neutrophils (IC50 = 0.4 ± 0.1 μM), however was ineffective on isolated 5-LOX. We exclude an interference with the 5-LOX-activating protein (FLAP) in HEK_5-LOX/±FLAP cells and suggest global effects on intact immune cells. Using LC-MS based targeted oxylipin metabolomics, we analyzed the effects of 5-LOX-inhibiting polyphenols on all branches of the AA cascade in Ca2+-ionophore-challenged neutrophils. While ε-viniferin causes a clear substrate shunt towards the remaining AA cascade enzymes (15-LOX, cyclooxygenase - COX-1/2, cytochrome P450), resveratrol inhibited the COX-1/2 pathway and showed a weak attenuation of 12/15-LOX activity. IRA had no impact on 15-LOX activity, but elevated the formation of COX-derived prostaglandins, having no inhibitory effects on COX-1/2. Overall, we show that food polyphenols have the ability to block 5-LOX activity and the oxylipin pattern is modulated with a remarkable compound/structural specificity. Taken the importance of polyphenols for a healthy diet and their concentration in food supplements into account, this finding justifies further investigation.
Collapse
Affiliation(s)
- Nicole M Hartung
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany; Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119 Wuppertal, Germany
| | - Jana Fischer
- Institute of Pharmacy, Friedrich-Schiller-University, Philosophenweg 14, 07743 Jena, Germany
| | - Annika I Ostermann
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany; Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119 Wuppertal, Germany
| | - Ina Willenberg
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany; Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119 Wuppertal, Germany
| | - Katharina M Rund
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany; Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119 Wuppertal, Germany
| | - Nils Helge Schebb
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, 30173 Hannover, Germany; Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119 Wuppertal, Germany
| | - Ulrike Garscha
- Institute of Pharmacy, Friedrich-Schiller-University, Philosophenweg 14, 07743 Jena, Germany.
| |
Collapse
|
29
|
Resveratrol as a Tumor-Suppressive Nutraceutical Modulating Tumor Microenvironment and Malignant Behaviors of Cancer. Int J Mol Sci 2019; 20:ijms20040925. [PMID: 30791624 PMCID: PMC6412705 DOI: 10.3390/ijms20040925] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor-suppressive effects of resveratrol have been shown in various types of cancer. However, regulation of tumor microenvironment by resveratrol is still unclear. Recent findings suggest resveratrol can potentiate its tumor-suppressive effect through modulation of the signaling pathways of cellular components (fibroblasts, macrophages and T cells). Also, studies have shown that resveratrol can suppress malignant phenotypes of cancer cells acquired in response to stresses of the tumor microenvironment, such as hypoxia, oxidative stress and inflammation. We discuss the effects of resveratrol on cancer cells in stress environment of tumors as well as interactions between cancer cells and non-cancer cells in this review.
Collapse
|
30
|
Schmidtke L, Schrick K, Saurin S, Käfer R, Gather F, Weinmann-Menke J, Kleinert H, Pautz A. The KH-type splicing regulatory protein (KSRP) regulates type III interferon expression post-transcriptionally. Biochem J 2019; 476:333-352. [PMID: 30578289 DOI: 10.1042/bcj20180522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Type III interferons (IFNs) are the latest members of the IFN family. They play an important role in immune defense mechanisms, especially in antiviral responses at mucosal sites. Moreover, they control inflammatory reactions by modulating neutrophil and dendritic cell functions. Therefore, it is important to identify cellular mechanisms involved in the control of type III IFN expression. All IFN family members contain AU-rich elements (AREs) in the 3'-untranslated regions (3'-UTR) of their mRNAs that determine mRNA half-life and consequently the expressional level of these cytokines. mRNA stability is controlled by different proteins binding to these AREs leading to either stabilization or destabilization of the respective target mRNA. The KH-type splicing regulatory protein KSRP (also named KHSRP) is an important negative regulator of ARE-containing mRNAs. Here, we identify the interferon lambda 3 (IFNL3) mRNA as a new KSRP target by pull-down and immunoprecipitation experiments, as well as luciferase reporter gene assays. We characterize the KSRP-binding site in the IFNL3 3'-UTR and demonstrate that KSRP regulates the mRNA half-life of the IFNL3 transcript. In addition, we detect enhanced expression of IFNL3 mRNA in KSRP-/- mice, establishing a negative regulatory function of KSRP in type III IFN expression also in vivo Besides KSRP the RNA-binding protein AUF1 (AU-rich element RNA-binding protein 1) also seems to be involved in the regulation of type III IFN mRNA expression.
Collapse
Affiliation(s)
- Lisa Schmidtke
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | - Katharina Schrick
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | - Sabrina Saurin
- First Medical Department, University Medical Center of the Johannes Gutenberg-University, Langenbeck Str. 1, 55101 Mainz, Germany
| | - Rudolf Käfer
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | - Fabian Gather
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | - Julia Weinmann-Menke
- First Medical Department, University Medical Center of the Johannes Gutenberg-University, Langenbeck Str. 1, 55101 Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Str. 67, 55101 Mainz, Germany
| |
Collapse
|
31
|
D'Agostino VG, Sighel D, Zucal C, Bonomo I, Micaelli M, Lolli G, Provenzani A, Quattrone A, Adami V. Screening Approaches for Targeting Ribonucleoprotein Complexes: A New Dimension for Drug Discovery. SLAS DISCOVERY 2019; 24:314-331. [PMID: 30616427 DOI: 10.1177/2472555218818065] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RNA-binding proteins (RBPs) are pleiotropic factors that control the processing and functional compartmentalization of transcripts by binding primarily to mRNA untranslated regions (UTRs). The competitive and/or cooperative interplay between RBPs and an array of coding and noncoding RNAs (ncRNAs) determines the posttranscriptional control of gene expression, influencing protein production. Recently, a variety of well-recognized and noncanonical RBP domains have been revealed by modern system-wide analyses, underlying an evolving classification of ribonucleoproteins (RNPs) and their importance in governing physiological RNA metabolism. The possibility of targeting selected RNA-protein interactions with small molecules is now expanding the concept of protein "druggability," with new implications for medicinal chemistry and for a deeper characterization of the mechanism of action of bioactive compounds. Here, taking SF3B1, HuR, LIN28, and Musashi proteins as paradigmatic case studies, we review the strategies applied for targeting RBPs, with emphasis on the technological advancements to study protein-RNA interactions and on the requirements of appropriate validation strategies to parallel high-throughput screening (HTS) efforts.
Collapse
Affiliation(s)
- Vito Giuseppe D'Agostino
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Denise Sighel
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Chiara Zucal
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Isabelle Bonomo
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Mariachiara Micaelli
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Graziano Lolli
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Alessandro Provenzani
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Alessandro Quattrone
- 1 University of Trento, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| | - Valentina Adami
- 2 University of Trento, HTS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), Trento, Italy
| |
Collapse
|
32
|
Bros M, Youns M, Kollek V, Buchmüller D, Bollmann F, Seo EJ, Schupp J, Montermann E, Usanova S, Kleinert H, Efferth T, Reske-Kunz AB. Differentially Tolerized Mouse Antigen Presenting Cells Share a Common miRNA Signature Including Enhanced mmu-miR-223-3p Expression Which Is Sufficient to Imprint a Protolerogenic State. Front Pharmacol 2018; 9:915. [PMID: 30174602 PMCID: PMC6108336 DOI: 10.3389/fphar.2018.00915] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are pivotal for the induction and maintenance of antigen-specific tolerance and immunity. miRNAs mediate post-transcriptional gene regulation and control in part the differentiation and stimulation-induced immunogenic function of DCs. However, the relevance of miRNAs for the induction and maintenance of a tolerogenic state of DCs has scarcely been highlighted yet. We differentiated mouse bone marrow cells to conventional/myeloid DCs or to tolerogenic antigen presenting cells (APCs) by using a glucocorticoid (dexamethasone) or interleukin-10, and assessed the miRNA expression patterns of unstimulated and LPS-stimulated cell populations by array analysis and QPCR. Differentially tolerized mouse APCs convergingly down-regulated a set of miRNA species at either state of activation as compared with the corresponding control DC population (mmu-miR-9-5p, mmu-miR-9-3p, mmu-miR-155-5p). These miRNAs were also upregulated in control DCs in response to stimulation. In contrast, miRNAs that were convergingly upregulated in both tolerized APC groups at stimulated state (mmu-miR-223-3p, mmu-miR-1224-5p) were downregulated in control DCs in response to stimulation. Overexpression of mmu-miR-223-3p in DCs was sufficient to prevent stimulation-associated acquisition of potent T cell stimulatory capacity. Overexpression of mmu-miR-223-3p in a DC line resulted in attenuated expression of known (Cflar, Rasa1, Ras) mRNA targets of this miRNA species shown to affect pathways that control DC activation. Taken together, we identified sets of miRNAs convergingly regulated in differentially tolerized APCs, which may contribute to imprint stimulation-resistant tolerogenic function as demonstrated for mmu-miR-223-3p. Knowledge of miRNAs with protolerogenic function enables immunotherapeutic approaches aimed to modulate immune responses by regulating miRNA expression.
Collapse
Affiliation(s)
- Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Mahmoud Youns
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Verena Kollek
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Diana Buchmüller
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Franziska Bollmann
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ean-Jeong Seo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Svetlana Usanova
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Angelika B Reske-Kunz
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
33
|
Li L, Qiu RL, Lin Y, Cai Y, Bian Y, Fan Y, Gao XJ. Resveratrol suppresses human cervical carcinoma cell proliferation and elevates apoptosis via the mitochondrial and p53 signaling pathways. Oncol Lett 2018; 15:9845-9851. [PMID: 29928358 PMCID: PMC6004645 DOI: 10.3892/ol.2018.8571] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have demonstrated the apoptotic and anti-proliferative effects of resveratrol, a natural polyphenolic phytoalexin, on various cancer cell lines. However, the effects of resveratrol on the regulation of human cervical carcinoma, and the mechanisms underlying these effects, remain to be elucidated. In the present study, the potential mechanisms underlying the effects of resveratrol in HeLa cervical carcinoma cells were investigated. The results revealed that resveratrol inhibited proliferation and induced apoptosis in HeLa human cervical cancer cells in a dose-dependent and time-dependent manner. Resveratrol induced cell shrinkage in HeLa cells and apoptosis accompanied by the activation of caspase-3 and -9. Furthermore, resveratrol upregulated the expression of the pro-apoptotic B-cell lymphoma (Bcl)-2-associated X protein and downregulated the expression of the anti-apoptotic proteins Bcl-2 and Bcl-extra large in HeLa cells. In addition, p53, a protein that is essential for cell survival and cell cycle progression, exhibited elevated expression levels in resveratrol-treated HeLa cells. Therefore, resveratrol may be a promising novel inhibitor of human cervical cancer.
Collapse
Affiliation(s)
- Lin Li
- Institute of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Rong-Li Qiu
- Institute of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yu Lin
- CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Ying Cai
- Nanjing General Hospital of Nanjing Military Command, Nanjing, Jiangsu 210002, P.R. China
| | - Yong Bian
- Institute of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yan Fan
- Institute of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xiao-Jun Gao
- Institute of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
34
|
1,8-cineole prevents UVB-induced skin carcinogenesis by targeting the aryl hydrocarbon receptor. Oncotarget 2017; 8:105995-106008. [PMID: 29285309 PMCID: PMC5739696 DOI: 10.18632/oncotarget.22519] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022] Open
Abstract
1,8-cineole is a natural monoterpene cyclic ether present in Eucalyptus, and has been reported to exhibit anti-inflammatory and antioxidant effects. However, the preventive effect of 1,8-cineole on skin carcinogenesis and the molecular mechanism of action responsible remains unknown. In the present study, we investigated the effect of 1,8-cineole on UVB-induced skin carcinogenesis. 1,8-cineole inhibited UVB-induced cyclooxygenase-2 (COX-2) protein and mRNA expression and prostaglandin E2 (PGE2) generation in HaCaT cells. 1,8-cineole also inhibited phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, and phosphorylation of its upstream kinases, c-Src and epidermal growth factor receptor (EGFR). Quantitative real-time RT-PCR (qRT-PCR) and drug affinity responsive target stability (DARTS) assay results showed that 1,8-cineole suppressed UVB-induced expression of a target gene of the aryl hydrocarbon receptor (AhR), cyp1a1, and directly binds to AhR. Knockdown of AhR suppressed COX-2 expression as well as phosphorylation of ERK1/2 in HaCaT cells. Furthermore, topical treatment of 1,8-cineole on mouse skin delayed tumor incidence and reduced tumor numbers, while inhibiting COX-2 expression in vivo. Taken together, these results suggest that 1,8-cineole is a potent chemopreventive agent that inhibits UVB-induced COX-2 expression by targeting AhR to suppress UVB-induced skin carcinogenesis.
Collapse
|
35
|
Wu S, Huang J. Resveratrol alleviates Staphylococcus aureus pneumonia by inhibition of the NLRP3 inflammasome. Exp Ther Med 2017; 14:6099-6104. [PMID: 29285164 DOI: 10.3892/etm.2017.5337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 04/10/2017] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus (S. aureus) causes a wide variety of infections, including S. aureus pneumonia. Resveratrol, a natural polyphenolic compound contained in several plant species, exerts various activities, including anti-bacterial and pulmonary protective activities. The aim of the present study was to evaluate the protective effect of resveratrol on a murine model of S. aureus pneumonia and to elucidate the underlying mechanisms. It was found that resveratrol significantly reduced S. aureus-induced mortality, ameliorated lung injury and decreased cytokine levels in the bronchoalveolar lavage fluid and lung tissue of S. aureus infected-mice. In addition, reverse-transcription quantitative polymerase chain reaction and western blot assays showed that resveratrol markedly decreased the mRNA and protein expression of nucleotide-binding domain and leucine-rich repeat containing gene family pyrin domain containing 3 protein (NLRP3), apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain and caspase-1. These results demonstrated that resveratrol significantly alleviates S. aureus pneumonia in mice, the possible underlying mechanism of which is associated with the inhibition of the NLRP3 inflammasome. The present study suggested that resveratrol is a potentially useful agent for the treatment of S. aureus pneumonia and S. aureus-induced infectious diseases.
Collapse
Affiliation(s)
- Suxia Wu
- Department of Respiratory Medicine, Zhangjiagang City Jingfeng People's Hospital, Suzhou, Jiangsu 215625, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
36
|
THP-1-derived macrophages render lung epithelial cells hypo-responsive to Legionella pneumophila - a systems biology study. Sci Rep 2017; 7:11988. [PMID: 28931863 PMCID: PMC5607273 DOI: 10.1038/s41598-017-12154-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/04/2017] [Indexed: 11/25/2022] Open
Abstract
Immune response in the lung has to protect the huge alveolar surface against pathogens while securing the delicate lung structure. Macrophages and alveolar epithelial cells constitute the first line of defense and together orchestrate the initial steps of host defense. In this study, we analysed the influence of macrophages on type II alveolar epithelial cells during Legionella pneumophila-infection by a systems biology approach combining experimental work and mathematical modelling. We found that L. pneumophila-infected THP-1-derived macrophages provoke a pro-inflammatory activation of neighboring lung epithelial cells, but in addition render them hypo-responsive to direct infection with the same pathogen. We generated a kinetic mathematical model of macrophage activation and identified a paracrine mechanism of macrophage-secreted IL-1β inducing a prolonged IRAK-1 degradation in lung epithelial cells. This intercellular crosstalk may help to avoid an overwhelming inflammatory response by preventing excessive local secretion of pro-inflammatory cytokines and thereby negatively regulating the recruitment of immune cells to the site of infection. This suggests an important but ambivalent immunomodulatory role of macrophages in lung infection.
Collapse
|
37
|
Gan Z, Powell FL, Zambon AC, Buchholz KS, Fu Z, Ocorr K, Bodmer R, Moya EA, Stowe JC, Haddad GG, McCulloch AD. Transcriptomic analysis identifies a role of PI3K-Akt signalling in the responses of skeletal muscle to acute hypoxia in vivo. J Physiol 2017; 595:5797-5813. [PMID: 28688178 PMCID: PMC5577531 DOI: 10.1113/jp274556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/19/2017] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS Changes in gene expression that occur within hours of exposure to hypoxia in in vivo skeletal muscles remain unexplored. Two hours of hypoxia caused significant down-regulation of extracellular matrix genes followed by a shift at 6 h to altered expression of genes associated with the nuclear lumen while respiratory and blood gases were stabilized. Enrichment analysis of mRNAs classified by stability rates suggests an attenuation of post-transcriptional regulation within hours of hypoxic exposure, where PI3K-Akt signalling was suggested to have a nodal role by pathway analysis. Experimental measurements and bioinformatic analyses suggested that the dephosphorylation of Akt after 2 h of hypoxic exposure might deactivate RNA-binding protein BRF1, hence resulting in the selective degradation of mRNAs. ABSTRACT The effects of acute hypoxia have been widely studied, but there are few studies of transcriptional responses to hours of hypoxia in vivo, especially in hypoxia-tolerant tissues like skeletal muscles. We used RNA-seq to analyse gene expression in plantaris muscles while monitoring respiration, arterial blood gases, and blood glucose in mice exposed to 8% O2 for 2 or 6 h. Rapid decreases in blood gases and a slower reduction in blood glucose suggest stress, which was accompanied by widespread changes in gene expression. Early down-regulation of genes associated with the extracellular matrix was followed by a shift to genes associated with the nuclear lumen. Most of the early down-regulated genes had mRNA half-lives longer than 2 h, suggesting a role for post-transcriptional regulation. These transcriptional changes were enriched in signalling pathways in which the PI3K-Akt signalling pathway was identified as a hub. Our analyses indicated that gene targets of PI3K-Akt but not HIF were enriched in early transcriptional responses to hypoxia. Among the PI3K-Akt targets, 75% could be explained by a deactivation of adenylate-uridylate-rich element (ARE)-binding protein BRF1, a target of PI3K-Akt. Consistent decreases in the phosphorylation of Akt and BRF1 were experimentally confirmed following 2 h of hypoxia. These results suggest that the PI3K-Akt signalling pathway might play a role in responses induced by acute hypoxia in skeletal muscles, partially through the dephosphorylation of ARE-binding protein BRF1.
Collapse
Affiliation(s)
- Zhuohui Gan
- School of Basic Medical SciencesWenzhou Medical UniversityWenzhou325035ZhejiangChina
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Frank L. Powell
- Department of MedicineUniversity of California San DiegoLa JollaCA92093USA
| | - Alexander C. Zambon
- Department of Biopharmaceutical SciencesKeck Graduate InstituteClaremontCA91711USA
| | - Kyle S. Buchholz
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Zhenxing Fu
- Department of MedicineUniversity of California San DiegoLa JollaCA92093USA
| | - Karen Ocorr
- Development, Aging and Regeneration ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCA92037USA
| | - Rolf Bodmer
- Development, Aging and Regeneration ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCA92037USA
| | - Esteban A. Moya
- Department of MedicineUniversity of California San DiegoLa JollaCA92093USA
| | - Jennifer C. Stowe
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Gabriel G. Haddad
- Department of PediatricsUniversity of California San DiegoLa JollaCA92093USA
- Department of NeurosciencesUniversity of California San DiegoLa JollaCA92093USA
- Rady Children's Hospital San Diego3020 Children's WaySan DiegoCA92123USA
| | - Andrew D. McCulloch
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Department of MedicineUniversity of California San DiegoLa JollaCA92093USA
| |
Collapse
|
38
|
Käfer R, Schrick K, Schmidtke L, Montermann E, Hobernik D, Bros M, Chen CY, Kleinert H, Pautz A. Inactivation of the KSRP gene modifies collagen antibody induced arthritis. Mol Immunol 2017; 87:207-216. [DOI: 10.1016/j.molimm.2017.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022]
|
39
|
Valdés A, García-Cañas V, Pérez-Sánchez A, Barrajón-Catalán E, Ruiz-Torres V, Artemenko KA, Micol V, Bergquist J, Cifuentes A. Shotgun proteomic analysis to study the decrease of xenograft tumor growth after rosemary extract treatment. J Chromatogr A 2017; 1499:90-100. [PMID: 28389096 DOI: 10.1016/j.chroma.2017.03.072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/23/2017] [Accepted: 03/25/2017] [Indexed: 12/18/2022]
Abstract
The antiproliferative activity of Rosemary (Rosmarinus officinalis) has been widely studied in different in vitro and in vivo models, which demonstrate that rosemary extracts inhibit the cellular proliferation due to its ability to interact with a wide spectrum of molecular targets. However, a comprehensive proteomics study in vivo has not been carried out yet. In the present work, the effects of rosemary extract on xenograft tumor growth has been studied and, for the first time, a shotgun proteomic analysis based on nano-LC-MS/MS together with stable isotope dimethyl labeling (DML) has been applied to investigate the global protein changes in vivo. Our results show that the daily administration of a polyphenol-enriched rosemary extract reduces the progression of colorectal cancer in vivo with the subsequent deregulation of 74 proteins. The bioinformatic analysis of these proteins indicates that the rosemary extract mainly alters the RNA Post-Transcriptional Modification, the Protein Synthesis and the Amino Acid Metabolism functions and suggests the inactivation of the oncogene MYC. These results demonstrate the high utility of the proposed analytical methodology to determine, simultaneously, the expression levels of a large number of protein biomarkers and to generate new hypothesis about the molecular mechanisms of this extract in vivo.
Collapse
Affiliation(s)
- Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC), Nicolas Cabrera 9, 28049, Madrid, Spain
| | - Virginia García-Cañas
- Molecular Nutrition and Metabolism, Institute of Food Science Research (CIAL, CSIC), Nicolas Cabrera 9, 28049 Madrid, Spain
| | - Almudena Pérez-Sánchez
- Institute of Molecular and Cellular Biology, Miguel Hernández University, Avda. Universidad s/n, Elche 03202, Spain
| | - Enrique Barrajón-Catalán
- Institute of Molecular and Cellular Biology, Miguel Hernández University, Avda. Universidad s/n, Elche 03202, Spain
| | - Verónica Ruiz-Torres
- Institute of Molecular and Cellular Biology, Miguel Hernández University, Avda. Universidad s/n, Elche 03202, Spain
| | - Konstantin A Artemenko
- Analytical Chemistry, Department of Chemistry-BMC, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden
| | - Vicente Micol
- Institute of Molecular and Cellular Biology, Miguel Hernández University, Avda. Universidad s/n, Elche 03202, Spain; CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), Spain
| | - Jonas Bergquist
- Analytical Chemistry, Department of Chemistry-BMC, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research (CIAL, CSIC), Nicolas Cabrera 9, 28049, Madrid, Spain.
| |
Collapse
|
40
|
Bikkavilli RK, Zerayesus SA, Van Scoyk M, Wilson L, Wu PY, Baskaran A, Tang K, Raheem S, Samuelson BA, Reddy NM, Reddy SP, Cool CD, Kosmider B, Avasarala S, Winn RA. K-homology splicing regulatory protein (KSRP) promotes post-transcriptional destabilization of Spry4 transcripts in non-small cell lung cancer. J Biol Chem 2017; 292:7423-7434. [PMID: 28275056 PMCID: PMC5418043 DOI: 10.1074/jbc.m116.757906] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 02/24/2017] [Indexed: 12/20/2022] Open
Abstract
AU-rich element-binding proteins (ARE-BPs) offer post-transcriptional regulation of gene expression via physical interaction and recruitment of RNA decay machinery to the AU-rich elements within the 3′-UTR of the target transcripts. However, the role of ARE-BPs in lung cancer remains poorly understood. In this study, we have identified that K-homology splicing regulatory protein (KSRP), an ARE-BP, is robustly up-regulated in human lung cancer. Importantly, Kaplan-Meier survival analysis indicated that elevated KSRP expression was correlated with poor overall survival of lung cancer patients. Furthermore, cigarette smoke, a leading risk factor for lung cancer, was also identified to be an important contributor to increased KSRP expression. Remarkably, silencing of KSRP decreased cell proliferation, reversed anchorage-independent growth, and reduced migration/invasion, suggesting an oncogenic role for KSRP in lung cancer. Finally, we provide mechanistic evidence that KSRP promotes the down-regulation of Spry4 by a previously unidentified mechanism, i.e. post-transcriptional mRNA regulation.
Collapse
Affiliation(s)
- Rama Kamesh Bikkavilli
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Sereke Adam Zerayesus
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Michelle Van Scoyk
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Lora Wilson
- Department of Pathology and Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Pei-Ying Wu
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Abhinaya Baskaran
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Ke Tang
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Syed Raheem
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Blain A Samuelson
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Narsa M Reddy
- Division of Developmental Biology and Basic Research, Department of Pediatrics,University of Illinois, Chicago, Illinois 60612
| | - Sekhar P Reddy
- Division of Developmental Biology and Basic Research, Department of Pediatrics,University of Illinois, Chicago, Illinois 60612
| | - Carlyne D Cool
- Department of Pathology and Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Beata Kosmider
- Departments of Physiology, Thoracic Medicine, and Surgery, Lewis Katz School of Medicine and.,Center for Inflammation, Translational, and Clinical Lung Research, Temple University, Philadelphia, Pennsylvania 19140.,Department of Medicine, National Jewish Health, Denver, Colorado 80206, and
| | - Sreedevi Avasarala
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and
| | - Robert A Winn
- From the Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and .,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
41
|
Resveratrol limits epithelial to mesenchymal transition through modulation of KHSRP/hnRNPA1-dependent alternative splicing in mammary gland cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:291-298. [PMID: 28088441 DOI: 10.1016/j.bbagrm.2017.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/29/2022]
Abstract
Resveratrol (RESV) is a natural polyphenolic compound endowed with anti-inflammatory, anti-proliferative, as well as pro-apoptotic activities that make it a potential anti-tumor compound. Here we show that RESV counteracts the TGF-β-induced Epithelial to Mesenchymal Transition (EMT) phenotype in mammary gland cells and affects the alternative exon usage of pre-mRNAs that encode crucial factors in adhesion and migration -including CD44, ENAH, and FGFR2- in a panel of immortalized and transformed mammary gland cells. RESV causes a shift from the mesenchymal-specific forms of these factors to the respective epithelial forms and increases the expression of the RNA-binding proteins KHSRP and hnRNPA1. From a mechanistic point of view, we show that the combined silencing of KHSRP and hnRNPA1 prevents the RESV-dependent inclusion of the epithelial-type exons in the Cd44 pre-mRNA. Our findings support an unexpected regulatory mechanism where RESV limits EMT by controlling gene expression at post-transcriptional level.
Collapse
|
42
|
Willson CM, Grundmann O. In vitro assays in natural products research - a matter of concentration and relevance to in vivo administration using resveratrol, α-mangostin/γ-mangostin and xanthohumol as examples. Nat Prod Res 2016; 31:492-506. [PMID: 27234135 DOI: 10.1080/14786419.2016.1190721] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Herbal or botanical dietary supplements are an ever increasingly popular category of products in the United States and around the world. In vitro data can provide meaningful insight into the potential target and mechanism of action for a proposed active compound but may also be misused to promote a supplement to consumers with unverified health claims. In vitro data need to be considered alongside pharmacokinetic and pharmacodynamic data in preclinical animal and clinical human trials. While considerable activity of compounds and extracts in vitro may lead to further testing in vivo, in many instances, concentrations tested in cell lines or isolated targets are not achievable at the target site in vivo. Thus, whether the in vitro data are relevant to humans after oral administration is questionable. This review will discuss this discrepancy using in vitro and in vivo data of resveratrol, xanthones (α-mangostin and γ-mangostin) and xanthohumol.
Collapse
Affiliation(s)
- C M Willson
- a Department of Medicinal Chemistry , College of Pharmacy, University of Florida , Gainesville , FL , USA
| | - O Grundmann
- a Department of Medicinal Chemistry , College of Pharmacy, University of Florida , Gainesville , FL , USA
| |
Collapse
|
43
|
Shin JS, Choi HE, Seo S, Choi JH, Baek NI, Lee KT. Berberine Decreased Inducible Nitric Oxide Synthase mRNA Stability through Negative Regulation of Human Antigen R in Lipopolysaccharide-Induced Macrophages. J Pharmacol Exp Ther 2016; 358:3-13. [PMID: 27189969 DOI: 10.1124/jpet.115.231043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022] Open
Abstract
Berberine, a major isoquinoline alkaloid found in medicinal herbs, has been reported to possess anti-inflammatory effects; however, the underlying mechanisms responsible for its actions are poorly understood. In the present study, we investigated the inhibitory effects of berberine and the molecular mechanisms involved in lipopolysaccharide (LPS)-treated RAW 264.7 and THP-1 macrophages and its effects in LPS-induced septic shock in mice. In both macrophage cell types, berberine inhibited the LPS-induced nitric oxide (NO) production and inducible NO synthase (iNOS) protein expression, but it had no effect on iNOS mRNA transcription. Suppression of LPS-induced iNOS protein expression by berberine occurred via a human antigen R (HuR)-mediated reduction of iNOS mRNA stability. Molecular data revealed that the suppression on the LPS-induced HuR binding to iNOS mRNA by berberine was accompanied by a reduction in nucleocytoplasmic HuR shuttling. Pretreatment with berberine reduced LPS-induced iNOS protein expression and the cytoplasmic translocation of HuR in liver tissues and increased the survival rate of mice with LPS-induced endotoxemia. These results show that the suppression of iNOS protein expression by berberine under LPS-induced inflammatory conditions is associated with a reduction in iNOS mRNA stability resulting from inhibition of the cytoplasmic translocation of HuR.
Collapse
Affiliation(s)
- Ji-Sun Shin
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| | - Hye-Eun Choi
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| | - SeungHwan Seo
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| | - Jung-Hye Choi
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| | - Nam-In Baek
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry (J.-S.S., H.-E.C., SH.S., K.-T.L.), Department of Life and Nanopharmaceutical Science (H.-E.C., SH.S., J.-H.C.,K.-T. L), and Department of Oriental Pharmaceutical Science, College of Pharmacy (J.-H.C.), Kyung Hee University, Seoul, Republic of Korea; and Graduate School of Biotechnology & Plant Metabolism Research Center, Kyung Hee University, Suwon, Republic of Korea (N.-I.B.)
| |
Collapse
|
44
|
Xia N, Daiber A, Förstermann U, Li H. Antioxidant effects of resveratrol in the cardiovascular system. Br J Pharmacol 2016; 174:1633-1646. [PMID: 27058985 DOI: 10.1111/bph.13492] [Citation(s) in RCA: 401] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 03/16/2016] [Accepted: 03/31/2016] [Indexed: 01/04/2023] Open
Abstract
The antioxidant effects of resveratrol (3,5,4'-trihydroxy-trans-stilbene) contribute substantially to the health benefits of this compound. Resveratrol has been shown to be a scavenger of a number of free radicals. However, the direct scavenging activities of resveratrol are relatively poor. The antioxidant properties of resveratrol in vivo are more likely to be attributable to its effect as a gene regulator. Resveratrol inhibits NADPH oxidase-mediated production of ROS by down-regulating the expression and activity of the oxidase. This polyphenolic compound reduces mitochondrial superoxide generation by stimulating mitochondria biogenesis. Resveratrol prevents superoxide production from uncoupled endothelial nitric oxide synthase by up-regulating the tetrahydrobiopterin-synthesizing enzyme GTP cyclohydrolase I. In addition, resveratrol increases the expression of various antioxidant enzymes. Some of the gene-regulating effects of resveratrol are mediated by the histone/protein deacetylase sirtuin 1 or by the nuclear factor-E2-related factor-2. In this review article, we have also summarized the cardiovascular effects of resveratrol observed in clinical trials. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Andreas Daiber
- 2nd Medical Department, Cardiology and Angiology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
45
|
Wang Z, Gu Z, Shen Y, Wang Y, Li J, Lv H, Huo K. The Natural Product Resveratrol Inhibits Yeast Cell Separation by Extensively Modulating the Transcriptional Landscape and Reprogramming the Intracellular Metabolome. PLoS One 2016; 11:e0150156. [PMID: 26950930 PMCID: PMC4780762 DOI: 10.1371/journal.pone.0150156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/10/2016] [Indexed: 12/28/2022] Open
Abstract
An increasing number of studies have shown that the promising compound resveratrol treats multiple diseases, such as cancer and aging; however, the resveratrol mode-of-action (MoA) remains largely unknown. Here, by virtue of multiple omics approaches, we adopted fission yeast as a model system with the goal of dissecting the common MoA of the anti-proliferative activity of resveratrol. We found that the anti-proliferative activity of resveratrol is mainly due to its unique role of inhibiting the separation of sister cells, similar phenotype with the C2H2 zinc finger transcription factor Ace2 knock-out strain. Microarray analysis shown that resveratrol has extensive impact on the fission yeast transcription levels. Among the changed gene's list, 40% of up-regulated genes are Core Environmental Stress Responses genes, and 57% of the down-regulated genes are periodically expressed. Moreover, resveratrol leverages the metabolome, which unbalances the intracellular pool sizes of several classes of amino acids, nucleosides, sugars and lipids, thus reflecting the remodulated metabolic networks. The complexity of the resveratrol MoA displayed in previous reports and our work demonstrates that multiple omics approaches must be applied together to obtain a complete picture of resveratrol's anti-proliferative function.
Collapse
Affiliation(s)
- Zhe Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Song-Hu Road, Shanghai, 200438, China
- Division of Infectious Diseases, Weill Medical College of Cornell University, 413 E 69th St, New York, NY, 10021, United States of America
- * E-mail: (KH); (ZW); (HL)
| | - Zhongkai Gu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Song-Hu Road, Shanghai, 200438, China
- Institutes of Biomedical Sciences, Fudan University, 130 Dong-An Road, Shanghai, 200032, China
| | - Yan Shen
- Institutes of Biomedical Sciences, Fudan University, 130 Dong-An Road, Shanghai, 200032, China
| | - Yang Wang
- Institutes of Biomedical Sciences, Fudan University, 130 Dong-An Road, Shanghai, 200032, China
| | - Jing Li
- Department of Biological Sciences and Biotechnology, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, 100084, China
| | - Hong Lv
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Song-Hu Road, Shanghai, 200438, China
- * E-mail: (KH); (ZW); (HL)
| | - Keke Huo
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, 2005 Song-Hu Road, Shanghai, 200438, China
- * E-mail: (KH); (ZW); (HL)
| |
Collapse
|
46
|
Murat N, Korhan P, Kizer O, Evcim S, Kefi A, Demir Ö, Gidener S, Atabey N, Esen AA. Resveratrol Protects and Restores Endothelium-Dependent Relaxation in Hypercholesterolemic Rabbit Corpus Cavernosum. J Sex Med 2016; 13:12-21. [DOI: 10.1016/j.jsxm.2015.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/14/2015] [Indexed: 12/16/2022]
|
47
|
Briata P, Bordo D, Puppo M, Gorlero F, Rossi M, Perrone-Bizzozero N, Gherzi R. Diverse roles of the nucleic acid-binding protein KHSRP in cell differentiation and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 7:227-40. [PMID: 26708421 DOI: 10.1002/wrna.1327] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 12/15/2022]
Abstract
The single-stranded nucleic acid-binding protein KHSRP (KH-type splicing regulatory protein) modulates RNA life and gene expression at various levels. KHSRP controls important cellular functions as different as proliferation, differentiation, metabolism, and response to infectious agents. We summarize and discuss experimental evidence providing a potential link between changes in KHSRP expression/function and human diseases including neuromuscular disorders, obesity, type II diabetes, and cancer.
Collapse
Affiliation(s)
- Paola Briata
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Domenico Bordo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Margherita Puppo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Franco Gorlero
- S.C. Ginecologia e Ostetricia Galliera Hospital, Genova, Italy.,School of Medicine, DINOGMI, University of Genova, Genova, Italy
| | - Martina Rossi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Nora Perrone-Bizzozero
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Roberto Gherzi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|
48
|
D'Agostino VG, Lal P, Mantelli B, Tiedje C, Zucal C, Thongon N, Gaestel M, Latorre E, Marinelli L, Seneci P, Amadio M, Provenzani A. Dihydrotanshinone-I interferes with the RNA-binding activity of HuR affecting its post-transcriptional function. Sci Rep 2015; 5:16478. [PMID: 26553968 PMCID: PMC4639722 DOI: 10.1038/srep16478] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/14/2015] [Indexed: 01/07/2023] Open
Abstract
Post-transcriptional regulation is an essential determinant of gene expression programs in physiological and pathological conditions. HuR is a RNA-binding protein that orchestrates the stabilization and translation of mRNAs, critical in inflammation and tumor progression, including tumor necrosis factor-alpha (TNF). We identified the low molecular weight compound 15,16-dihydrotanshinone-I (DHTS), well known in traditional Chinese medicine practice, through a validated high throughput screening on a set of anti-inflammatory agents for its ability to prevent HuR:RNA complex formation. We found that DHTS interferes with the association step between HuR and the RNA with an equilibrium dissociation constant in the nanomolar range in vitro (Ki = 3.74 ± 1.63 nM). In breast cancer cell lines, short term exposure to DHTS influences mRNA stability and translational efficiency of TNF in a HuR-dependent manner and also other functional readouts of its post-transcriptional control, such as the stability of selected pre-mRNAs. Importantly, we show that migration and sensitivity of breast cancer cells to DHTS are modulated by HuR expression, indicating that HuR is among the preferential intracellular targets of DHTS. Here, we disclose a previously unrecognized molecular mechanism exerted by DHTS, opening new perspectives to therapeutically target the HuR mediated, post-transcriptional control in inflammation and cancer cells.
Collapse
Affiliation(s)
| | - Preet Lal
- Centre For Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Barbara Mantelli
- Centre For Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Christopher Tiedje
- Department of Biochemistry, Hannover Medical University, Hannover, D-30625, Germany
| | - Chiara Zucal
- Centre For Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Natthakan Thongon
- Centre For Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Matthias Gaestel
- Department of Biochemistry, Hannover Medical University, Hannover, D-30625, Germany
| | - Elisa Latorre
- Centre For Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Luciana Marinelli
- Department of Pharmacy, University of Naples "Federico II", Naples, 80131, Italy
| | | | - Marialaura Amadio
- Department of Drug Sciences, University of Pavia, Pavia, 27100, Italy
| | | |
Collapse
|
49
|
Riebel M, Sabel A, Claus H, Fronk P, Xia N, Li H, König H, Decker H. Influence of Laccase and Tyrosinase on the Antioxidant Capacity of Selected Phenolic Compounds on Human Cell Lines. Molecules 2015; 20:17194-207. [PMID: 26393557 PMCID: PMC6332179 DOI: 10.3390/molecules200917194] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/25/2015] [Accepted: 09/11/2015] [Indexed: 11/17/2022] Open
Abstract
Polyphenolic compounds affect the color, odor and taste of numerous food products of plant origin. In addition to the visual and gustatory properties, they serve as radical scavengers and have antioxidant effects. Polyphenols, especially resveratrol in red wine, have gained increasing scientific and public interest due to their presumptive beneficial impact on human health. Enzymatic oxidation of phenolic compounds takes place under the influence of polyphenol oxidases (PPO), including tyrosinase and laccase. Several studies have demonstrated the radical scavenger effect of plants, food products and individual polyphenols in vitro, but, apart from resveratrol, such impact has not been proved in physiological test systems. Furthermore, only a few data exist on the antioxidant capacities of the enzymatic oxidation products of phenolic compounds generated by PPO. We report here first results about the antioxidant effects of phenolic substances, before and after oxidation by fungal model tyrosinase and laccase. In general, the common chemical 2,2-diphenyl-1-picrylhydrazyl assay and the biological tests using two different types of cell cultures (monocytes and endothelial cells) delivered similar results. The phenols tested showed significant differences with respect to their antioxidant activity in all test systems. Their antioxidant capacities after enzymatic conversion decreased or increased depending on the individual PPO used.
Collapse
Affiliation(s)
- Matthias Riebel
- Institute for Molecular Biophysics, Johannes Gutenberg University of Mainz, Jakob-Welder-Weg, 26, 55128 Mainz, Germany.
| | - Andrea Sabel
- Institute for Microbiology and Wine Research, Johannes Gutenberg University of Mainz, Johann-Joachim-Becher-Weg, 15, 55128 Mainz, Germany.
| | - Harald Claus
- Institute for Microbiology and Wine Research, Johannes Gutenberg University of Mainz, Johann-Joachim-Becher-Weg, 15, 55128 Mainz, Germany.
| | - Petra Fronk
- Institute for Molecular Biophysics, Johannes Gutenberg University of Mainz, Jakob-Welder-Weg, 26, 55128 Mainz, Germany.
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center of Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany.
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center of Mainz, Obere Zahlbacher Str. 67, 55131 Mainz, Germany.
| | - Helmut König
- Institute for Microbiology and Wine Research, Johannes Gutenberg University of Mainz, Johann-Joachim-Becher-Weg, 15, 55128 Mainz, Germany.
| | - Heinz Decker
- Institute for Molecular Biophysics, Johannes Gutenberg University of Mainz, Jakob-Welder-Weg, 26, 55128 Mainz, Germany.
| |
Collapse
|
50
|
Lepak A, Gutmann A, Kulmer ST, Nidetzky B. Creating a Water-Soluble Resveratrol-Based Antioxidant by Site-Selective Enzymatic Glucosylation. Chembiochem 2015; 16:1870-1874. [DOI: 10.1002/cbic.201500284] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Indexed: 11/10/2022]
|