1
|
Zhong C, Wang S, Xia L, Yang X, Fang L, Zhang X, Wang M, Zhao H, Wang G, Wu J, Guo R, Zhong M, Gohda E. The tubulin polymerization inhibitor gambogenic acid induces myelodysplastic syndrome cell apoptosis through upregulation of Fas expression mediated by the NF-κB signaling pathway. Cancer Biol Ther 2024; 25:2427374. [PMID: 39540618 PMCID: PMC11572293 DOI: 10.1080/15384047.2024.2427374] [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: 06/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The development of an effective treatment for myelodysplastic syndrome (MDS) is needed due to the insufficient efficacy of current therapies. Gambogenic acid (GNA) is a xanthone constituent of gamboge, a resin secreted by Garcinia hanburyi Hook. f. GNA exhibits antitumor and apoptosis-inducing activities against some cancer cells, but the mechanism is unknown. This study aimed to validate the anti-proliferative and apoptosis-inducing effects of GNA on MDS cells and to elucidate the mechanisms underlying those activities. Apoptosis, proliferation and cell cycle of MDS-L cells were assessed by the caspase 3/7 assay, cell counting and flow cytometry, respectively. The levels of apoptotic, tubulin, NF-κB pathways, and Fas proteins were determined by Western blotting. CRISPR/Cas9 knockout (KO) plasmids were used to generate KO cells of p65 and Fas. MDS cell growth in a xenograft model was evaluated by the AkaBLI system. GNA induced MDS cell apoptosis, accompanied by a reduction in the anti-apoptotic protein MCL-1 expression, and inhibited their growth in vitro and in vivo. GNA combined with the MCL-1 inhibitor MIK665 potently suppressed the proliferation of MDS cells. GNA interfered with tubulin polymerization, resulting in G2/M arrest. GNA induced NF-κB activation and upregulation of Fas, the latter of which was inhibited by p65 KO. GNA-induced apoptosis was attenuated in either p65 KO or Fas KO cells. These results demonstrate that GNA inhibited tubulin polymerization and induced apoptosis of MDS cells through upregulation of Fas expression mediated by the NF-κB signaling pathway, suggesting a chemotherapeutic strategy for MDS by microtubule dynamics disruption.
Collapse
Affiliation(s)
- Cheng Zhong
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Postdoctoral Research Workstation of Integrated Traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shijun Wang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Postdoctoral Research Workstation of Integrated Traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Xia
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoman Yang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liguang Fang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianyi Zhang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengyue Wang
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Zhao
- Department of Pathology, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guanghui Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, Jining Medical University, Rizhao, China
| | - Jinglong Wu
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Ruijian Guo
- Department of Pharmaceutics, College of pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Zhong
- Department of Pharmacology and Toxicology, School of Pharmacy, Jining Medical University, Rizhao, China
| | - Eiichi Gohda
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
2
|
Morgan EL, Saleh AD, Cornelius S, Carlson SG, Toni T, Cheng H, Jeon J, Viswanathan R, Yang X, Silvin C, Clavijo PE, Sowers AL, Mitchell JB, Ormanoglu P, Lal Nag M, Martin SE, Chen Z, Van Waes C. Functional RNAi Screening Identifies G2/M and Kinetochore Components as Modulators of TNFα/NF-κB Prosurvival Signaling in Head and Neck Squamous Cell Carcinoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:2903-2918. [PMID: 39392349 PMCID: PMC11541648 DOI: 10.1158/2767-9764.crc-24-0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/14/2024] [Accepted: 10/09/2024] [Indexed: 10/12/2024]
Abstract
SIGNIFICANCE Here, RNAi library screening reveals that multiple G2/M and kinetochore components, including TTK/monopolar spindle 1, modulate TNFα-induced NF-κB activation, cell survival, and genotoxicity, underscoring their potential importance as therapeutic targets in HNSCC.
Collapse
Affiliation(s)
- Ethan L. Morgan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Anthony D. Saleh
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Shaleeka Cornelius
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Sophie G. Carlson
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Tiffany Toni
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Hui Cheng
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Jun Jeon
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Ramya Viswanathan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Xinping Yang
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Christopher Silvin
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Paul E. Clavijo
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Anastasia L. Sowers
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - James B. Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Pinar Ormanoglu
- RNAi Screening Facility, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Madhu Lal Nag
- RNAi Screening Facility, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Scott E. Martin
- RNAi Screening Facility, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
3
|
Mandal M, Rakib A, Mamun MAA, Kumar S, Park F, Hwang DJ, Li W, Miller DD, Singh UP. DJ-X-013 reduces LPS-induced inflammation, modulates Th17/ myeloid-derived suppressor cells, and alters NF-κB expression to ameliorate experimental colitis. Biomed Pharmacother 2024; 179:117379. [PMID: 39255739 PMCID: PMC11479677 DOI: 10.1016/j.biopha.2024.117379] [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: 06/20/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
SCOPE Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition of unknown etiology, although recent evidence suggests that it is caused by an excessive immune response to mucosal antigens. We determined the anti-inflammatory properties of novel compound DJ-X-013 in vitro in lipopolysaccharide (LPS)-induced macrophages and in an in vivo dextran sodium sulfate (DSS)-induced model of colitis. METHODS AND RESULTS To evaluate the anti-inflammatory properties of DJ-X-013, we used LPS-activated RAW 264.7 macrophages in vitro and a DSS-induced experimental model of colitis in vivo. We examine cellular morphology, and tissue architecture by histology, flow cytometry, RT-qPCR, multiplex, and immunoblot analysis to perform cellular and molecular studies. DJ-X-013 treatment altered cell morphology and expression of inflammatory cytokines in LPS-activated macrophages as compared to cells treated with LPS alone. DJ-X-013 also impeded the migration of RAW 264.7 macrophages by modulating cytoskeletal organization and suppressed the expression of NF-κB and inflammatory markers as compared to LPS alone. DJ-X-013 treatment improved body weight, and colon length and attenuated inflammation in the colon of DSS-induced colitis. Intriguingly, DSS-challenged mice treated with DJ-X-013 induced the numbers of myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), and natural killer T cells (NKT) in the colon lamina propria (LP) relative to DSS. DJ-X-013 also reduced the influx of neutrophils, TNF-α producing macrophages, restricted the number of Th17 cells, and suppressed inflammatory cytokines and NF-κB in the LP relative to DSS. CONCLUSION DJ-X-013 is proposed to be a therapeutic strategy for ameliorating inflammation and experimental colitis.
Collapse
Affiliation(s)
- Mousumi Mandal
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ahmed Rakib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Md Abdullah Al Mamun
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
4
|
Deswal B, Bagchi U, Santra MK, Garg M, Kapoor S. Inhibition of STAT3 by 2-Methoxyestradiol suppresses M2 polarization and protumoral functions of macrophages in breast cancer. BMC Cancer 2024; 24:1129. [PMID: 39256694 PMCID: PMC11389501 DOI: 10.1186/s12885-024-12871-w] [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: 08/05/2023] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Breast cancer metastasis remains the leading cause of cancer-related deaths in women worldwide. Infiltration of tumor-associated macrophages (TAMs) in the tumor stroma is known to be correlated with reduced overall survival. The inhibitors of TAMs are sought after for reprogramming the tumor microenvironment. Signal transducer and activator of transcription 3 (STAT3) is well known to contribute in pro-tumoral properties of TAMs. 2-Methoxyestradiol (2ME2), a potent anticancer and antiangiogenic agent, has been in clinical trials for treatment of breast cancer. Here, we investigated the potential of 2ME2 in modulating the pro-tumoral effects of TAMs in breast cancer. METHODS THP-1-derived macrophages were polarized to macrophages with or without 2ME2. The effect of 2ME2 on macrophage surface markers and anti-inflammatory genes was determined by Western blotting, flow cytometry, immunofluorescence, qRT‒PCR. The concentration of cytokines secreted by cells was monitored by ELISA. The effect of M2 macrophages on malignant properties of breast cancer cells was determined using colony formation, wound healing, transwell, and gelatin zymography assays. An orthotopic model of breast cancer was used to determine the effect of 2ME2 on macrophage polarization and metastasis in vivo. RESULTS First, our study found that polarization of monocytes to alternatively activated M2 macrophages is associated with the reorganization of the microtubule cytoskeleton. At lower concentrations, 2ME2 treatment depolymerized microtubules and reduced the expression of CD206 and CD163, suggesting that it inhibits the polarization of macrophages to M2 phenotype. However, the M1 polarization was not significantly affected at these concentrations. Importantly, 2ME2 inhibited the expression of several anti-inflammatory cytokines and growth factors, including CCL18, TGF-β, IL-10, FNT, arginase, CXCL12, MMP9, and VEGF-A, and hindered the metastasis-promoting effects of M2 macrophages. Concurrently, 2ME2 treatment reduced the expression of CD163 in tumors and inhibited lung metastasis in the orthotopic breast cancer model. Mechanistically, 2ME2 treatment reduced the phosphorylation and nuclear translocation of STAT3, an effect which was abrogated by colivelin. CONCLUSIONS Our study presents novel findings on mechanism of 2ME2 from the perspective of its effects on the polarization of the TAMs via the STAT3 signaling in breast cancer. Altogether, the data supports further clinical investigation of 2ME2 and its derivatives as therapeutic agents to modulate the tumor microenvironment and immune response in breast carcinoma.
Collapse
Affiliation(s)
- Bhawna Deswal
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India
| | - Urmi Bagchi
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India
| | - Manas Kumar Santra
- National Centre for Cell Science Complex, Savitribai Phule Pune University, Campus Ganeshkhind Road, Pune, Maharashtra, 411007, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India.
| | - Sonia Kapoor
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
5
|
Drif AI, Yücer R, Damiescu R, Ali NT, Abu Hagar TH, Avula B, Khan IA, Efferth T. Anti-Inflammatory and Cancer-Preventive Potential of Chamomile ( Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study. Biomedicines 2024; 12:1484. [PMID: 39062057 PMCID: PMC11275008 DOI: 10.3390/biomedicines12071484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIM Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer by focusing on its anti-inflammatory activity. METHODS AND RESULTS A virtual drug screening of 212 phytochemicals from chamomile revealed β-amyrin, β-eudesmol, β-sitosterol, apigenin, daucosterol, and myricetin as potent NF-κB inhibitors. The in silico results were verified through microscale thermophoresis, reporter cell line experiments, and flow cytometric determination of reactive oxygen species and mitochondrial membrane potential. An oncobiogram generated through comparison of 91 anticancer agents with known modes of action using the NCI tumor cell line panel revealed significant relationships of cytotoxic chamomile compounds, lupeol, and quercetin to microtubule inhibitors. This hypothesis was verified by confocal microscopy using α-tubulin-GFP-transfected U2OS cells and molecular docking of lupeol and quercetin to tubulins. Both compounds induced G2/M cell cycle arrest and necrosis rather than apoptosis. Interestingly, lupeol and quercetin were not involved in major mechanisms of resistance to established anticancer drugs (ABC transporters, TP53, or EGFR). Performing hierarchical cluster analyses of proteomic expression data of the NCI cell line panel identified two sets of 40 proteins determining sensitivity and resistance to lupeol and quercetin, further pointing to the multi-specific nature of chamomile compounds. Furthermore, lupeol, quercetin, and β-amyrin inhibited the mRNA expression of the proinflammatory cytokines IL-1β and IL6 in NF-κB reporter cells (HEK-Blue Null1). Moreover, Kaplan-Meier-based survival analyses with NF-κB as the target protein of these compounds were performed by mining the TCGA-based KM-Plotter repository with 7489 cancer patients. Renal clear cell carcinomas (grade 3, low mutational rate, low neoantigen load) were significantly associated with shorter survival of patients, indicating that these subgroups of tumors might benefit from NF-κB inhibition by chamomile compounds. CONCLUSION This study revealed the potential of chamomile, positioning it as a promising preventive agent against inflammation and cancer. Further research and clinical studies are recommended.
Collapse
Affiliation(s)
- Assia I. Drif
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Roxana Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Nadeen T. Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Tobias H. Abu Hagar
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Bharati Avula
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| |
Collapse
|
6
|
Liu Z, Nong K, Qin X, Fang X, Zhang B, Chen W, Wang Z, Wu Y, Shi H, Wang X, Liu Y, Guan Q, Zhang H. The antimicrobial peptide Abaecin alleviates colitis in mice by regulating inflammatory signaling pathways and intestinal microbial composition. Peptides 2024; 173:171154. [PMID: 38242174 DOI: 10.1016/j.peptides.2024.171154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
Abaecin is a natural antimicrobial peptide (AMP) rich in proline from bees. It is an important part of the innate humoral immunity of bees and has broad-spectrum antibacterial ability. This study aimed to determine the effect of Abaecin on dextran sulfate sodium (DSS) -induced ulcerative colitis (UC) in mice and to explore its related mechanisms. Twenty-four mice with similar body weight were randomly divided into 4 groups. 2.5% DSS was added to drinking water to induce colitis in mice. Abaecin and PBS were administered rectally on the third, fifth, and seventh days of the experimental period. The results showed that Abaecin significantly alleviated histological damage and intestinal mucosal barrier damage caused by colitis in mice, reduced the concentration of pro-inflammatory cytokines IL-1β, IL-6, TNF-α, IFN-γ, and the phosphorylation of NF-κB / MAPK inflammatory signaling pathway proteins, and improved the composition of intestinal microorganisms. These findings suggest that Abaecin may have potential prospects for the treatment of UC.
Collapse
Affiliation(s)
- Zhineng Liu
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Keyi Nong
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xinyun Qin
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xin Fang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Bin Zhang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Wanyan Chen
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Zihan Wang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Yijia Wu
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Huiyu Shi
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Xuemei Wang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China
| | - Youming Liu
- Yibin Academy of Agricultural Sciences, Yibin 644600, China
| | - Qingfeng Guan
- College of Life and Health, Hainan University, Haikou 570228, China
| | - Haiwen Zhang
- School of Tropical Agriculte and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
7
|
Arseni C, Samiotaki M, Panayotou G, Simos G, Mylonis I. Combinatorial regulation by ERK1/2 and CK1δ protein kinases leads to HIF-1α association with microtubules and facilitates its symmetrical distribution during mitosis. Cell Mol Life Sci 2024; 81:72. [PMID: 38300329 PMCID: PMC10834586 DOI: 10.1007/s00018-024-05120-7] [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: 10/30/2023] [Revised: 12/13/2023] [Accepted: 01/07/2024] [Indexed: 02/02/2024]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is the key transcriptional mediator of the cellular response to hypoxia and is also involved in cancer progression. Regulation of its oxygen-sensitive HIF-1α subunit involves post-translational modifications that control its stability, subcellular localization, and activity. We have previously reported that phosphorylation of the HIF-1α C-terminal domain by ERK1/2 promotes HIF-1α nuclear accumulation and stimulates HIF-1 activity while lack of this modification triggers HIF-1α nuclear export and its association with mitochondria. On the other hand, modification of the N-terminal domain of HIF-1α by CK1δ impairs HIF-1 activity by obstructing the formation of a HIF-1α/ARNT heterodimer. Investigation of these two antagonistic events by expressing double phospho-site mutants in HIF1A-/- cells under hypoxia revealed independent and additive phosphorylation effects that can create a gradient of HIF-1α subcellular localization and transcriptional activity. Furthermore, modification by CK1δ caused mitochondrial release of the non-nuclear HIF-1α form and binding to microtubules via its N-terminal domain. In agreement, endogenous HIF-1α could be shown to co-localize with mitotic spindle microtubules and interact with tubulin, both of which were inhibited by CK1δ silencing or inhibition. Moreover, CK1δ expression was necessary for equal partitioning of mother cell-produced HIF-1α to the daughter cell nuclei at the end of mitosis. Overall, our results suggest that phosphorylation by CK1δ stimulates the association of non-nuclear HIF-1α with microtubules, which may serve as a means to establish a symmetric distribution of HIF-1α during cell division under low oxygen conditions.
Collapse
Affiliation(s)
- Christina Arseni
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece
| | - Martina Samiotaki
- Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece
| | - George Panayotou
- Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece.
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada.
| | - Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece.
| |
Collapse
|
8
|
Wongsawat M, Glaharn S, Srisook C, Dechkhajorn W, Chaisri U, Punsawad C, Techarang T, Chotivanich K, Krudsood S, Viriyavejakul P. Immunofluorescence study of cytoskeleton in endothelial cells induced with malaria sera. Malar J 2024; 23:10. [PMID: 38183117 PMCID: PMC10770940 DOI: 10.1186/s12936-023-04833-7] [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: 07/10/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) play a major role in malaria pathogenesis, as a point of direct contact of parasitized red blood cells to the blood vessel wall. The study of cytoskeleton structures of ECs, whose main functions are to maintain shape and provide strength to the EC membrane is important in determining the severe sequelae of Plasmodium falciparum malaria. The work investigated the cytoskeletal changes (microfilaments-actin, microtubules-tubulin and intermediate filaments-vimentin) in ECs induced by malaria sera (Plasmodium vivax, uncomplicated P. falciparum and complicated P. falciparum), in relation to the levels of pro-inflammatory cytokines. METHODS Morphology and fluorescence intensity of EC cytoskeleton stimulated with malaria sera were evaluated using immunofluorescence technique. Levels of tumour necrosis factor (TNF) and interferon (IFN)-gamma (γ) were determined using enzyme-linked immunosorbent assay (ELISA). Control experimental groups included ECs incubated with media alone and non-malaria patient sera. Experimental groups consisted of ECs incubated with malaria sera from P. vivax, uncomplicated P. falciparum and complicated P. falciparum. Morphological scores of cytoskeletal alterations and fluorescence intensity were compared across each experiment group, and correlated with TNF and IFN-γ. RESULTS The four morphological changes of cytoskeleton included (1) shrinkage of cytoskeleton and ECs with cortical condensation, (2) appearance of eccentric nuclei, (3) presence of "spiking pattern" of cytoskeleton and EC membrane, and (4) fragmentation and discontinuity of cytoskeleton and ECs. Significant damages were noted in actin filaments compared to tubulin and vimentin filaments in ECs stimulated with sera from complicated P. falciparum malaria. Morphological damages to cytoskeleton was positively correlated with fluorescence intensity and the levels of TNF and IFN-γ. CONCLUSIONS ECs stimulated with sera from complicated P. falciparum malaria showed cytoskeletal alterations and increased in fluorescence intensity, which was associated with high levels of TNF and IFN-γ. Cytoskeletal changes of ECs incubated with complicated P. falciparum malaria sera can lead to EC junctional alteration and permeability changes, which is mediated through apoptotic pathway. The findings can serve as a basis to explore measures to strengthen EC cytoskeleton and alleviate severe malaria complications such as pulmonary oedema and cerebral malaria. In addition, immunofluorescence intensity of cytoskeleton could be investigated as potential prognostic indicator for malaria severity.
Collapse
Affiliation(s)
- Mathusorn Wongsawat
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Supattra Glaharn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Charit Srisook
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Wilanee Dechkhajorn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Tachpon Techarang
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Srivicha Krudsood
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand.
| |
Collapse
|
9
|
Mechanotransduction pathways in articular chondrocytes and the emerging role of estrogen receptor-α. Bone Res 2023; 11:13. [PMID: 36869045 PMCID: PMC9984452 DOI: 10.1038/s41413-023-00248-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/05/2022] [Accepted: 01/06/2023] [Indexed: 03/05/2023] Open
Abstract
In the synovial joint, mechanical force creates an important signal that influences chondrocyte behavior. The conversion of mechanical signals into biochemical cues relies on different elements in mechanotransduction pathways and culminates in changes in chondrocyte phenotype and extracellular matrix composition/structure. Recently, several mechanosensors, the first responders to mechanical force, have been discovered. However, we still have limited knowledge about the downstream molecules that enact alterations in the gene expression profile during mechanotransduction signaling. Recently, estrogen receptor α (ERα) has been shown to modulate the chondrocyte response to mechanical loading through a ligand-independent mechanism, in line with previous research showing that ERα exerts important mechanotransduction effects on other cell types, such as osteoblasts. In consideration of these recent discoveries, the goal of this review is to position ERα into the mechanotransduction pathways known to date. Specifically, we first summarize our most recent understanding of the mechanotransduction pathways in chondrocytes on the basis of three categories of actors, namely mechanosensors, mechanotransducers, and mechanoimpactors. Then, the specific roles played by ERα in mediating the chondrocyte response to mechanical loading are discussed, and the potential interactions of ERα with other molecules in mechanotransduction pathways are explored. Finally, we propose several future research directions that may advance our understanding of the roles played by ERα in mediating biomechanical cues under physiological and pathological conditions.
Collapse
|
10
|
Microtubules as a potential platform for energy transfer in biological systems: a target for implementing individualized, dynamic variability patterns to improve organ function. Mol Cell Biochem 2023; 478:375-392. [PMID: 35829870 DOI: 10.1007/s11010-022-04513-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
Variability characterizes the complexity of biological systems and is essential for their function. Microtubules (MTs) play a role in structural integrity, cell motility, material transport, and force generation during mitosis, and dynamic instability exemplifies the variability in the proper function of MTs. MTs are a platform for energy transfer in cells. The dynamic instability of MTs manifests itself by the coexistence of growth and shortening, or polymerization and depolymerization. It results from a balance between attractive and repulsive forces between tubulin dimers. The paper reviews the current data on MTs and their potential roles as energy-transfer cellular structures and presents how variability can improve the function of biological systems in an individualized manner. The paper presents the option for targeting MTs to trigger dynamic improvement in cell plasticity, regulate energy transfer, and possibly control quantum effects in biological systems. The described system quantifies MT-dependent variability patterns combined with additional personalized signatures to improve organ function in a subject-tailored manner. The platform can regulate the use of MT-targeting drugs to improve the response to chronic therapies. Ongoing trials test the effects of this platform on various disorders.
Collapse
|
11
|
Yim J, Lee J, Yi S, Koo JY, Oh S, Park H, Kim SS, Bae MA, Park J, Park SB. Phenotype-based screening rediscovered benzopyran-embedded microtubule inhibitors as anti-neuroinflammatory agents by modulating the tubulin-p65 interaction. Exp Mol Med 2022; 54:2200-2209. [PMID: 36509830 PMCID: PMC9743128 DOI: 10.1038/s12276-022-00903-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation is one of the critical processes implicated in central nervous system (CNS) diseases. Therefore, alleviating neuroinflammation has been highlighted as a therapeutic strategy for treating CNS disorders. However, the complexity of neuroinflammatory processes and poor drug transport to the brain are considerable hurdles to the efficient control of neuroinflammation using small-molecule therapeutics. Thus, there is a significant demand for new chemical entities (NCEs) targeting neuroinflammation. Herein, we rediscovered benzopyran-embedded tubulin inhibitor 1 as an anti-neuroinflammatory agent via phenotype-based screening. A competitive photoaffinity labeling study revealed that compound 1 binds to tubulin at the colchicine-binding site. Structure-activity relationship analysis of 1's analogs identified SB26019 as a lead compound with enhanced anti-neuroinflammatory efficacy. Mechanistic studies revealed that upregulation of the tubulin monomer was critical for the anti-neuroinflammatory activity of SB26019. We serendipitously found that the tubulin monomer recruits p65, inhibiting its translocation from the cytosol to the nucleus and blocking NF-κB-mediated inflammatory pathways. Further in vivo validation using a neuroinflammation mouse model demonstrated that SB26019 suppressed microglial activation by downregulating lba-1 and proinflammatory cytokines. Intraperitoneal administration of SB26019 showed its therapeutic potential as an NCE for successful anti-neuroinflammatory regulation. Along with the recent growing demands on tubulin modulators for treating various inflammatory diseases, our results suggest that colchicine-binding site-specific modulation of tubulins can be a potential strategy for preventing neuroinflammation and treating CNS diseases.
Collapse
Affiliation(s)
- Junhyeong Yim
- grid.31501.360000 0004 0470 5905Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826 Korea
| | - Jaeseok Lee
- grid.412010.60000 0001 0707 9039Department of Chemistry, Kangwon National University, Chuncheon, 24341 Korea
| | - Sihyeong Yi
- grid.31501.360000 0004 0470 5905CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826 Korea
| | - Ja Young Koo
- grid.31501.360000 0004 0470 5905CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826 Korea
| | - Sangmi Oh
- grid.31501.360000 0004 0470 5905CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826 Korea
| | - Hankum Park
- grid.31501.360000 0004 0470 5905CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826 Korea ,grid.31501.360000 0004 0470 5905Present Address: Department of Dental Sciences, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826 Korea
| | - Seong Soon Kim
- grid.29869.3c0000 0001 2296 8192Bio Platform Technology Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114 Korea
| | - Myung Ae Bae
- grid.29869.3c0000 0001 2296 8192Bio Platform Technology Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114 Korea ,grid.412786.e0000 0004 1791 8264Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon, 34114 Korea
| | - Jongmin Park
- grid.412010.60000 0001 0707 9039Department of Chemistry, Kangwon National University, Chuncheon, 24341 Korea ,grid.412010.60000 0001 0707 9039Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341 Korea
| | - Seung Bum Park
- grid.31501.360000 0004 0470 5905Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826 Korea ,grid.31501.360000 0004 0470 5905CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826 Korea
| |
Collapse
|
12
|
Schröder M, Petrova M, Vlahova Z, Dobrikov GM, Slavchev I, Pasheva E, Ugrinova I. In Vitro Anticancer Activity of Two Ferrocene-Containing Camphor Sulfonamides as Promising Agents against Lung Cancer Cells. Biomedicines 2022; 10:biomedicines10061353. [PMID: 35740374 PMCID: PMC9219647 DOI: 10.3390/biomedicines10061353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
The successful design of antitumour drugs often combines in one molecule different biologically active subunits that can affect various regulatory pathways in the cell and thus achieve higher efficacy. Two ferrocene derivatives, DK-164 and CC-78, with different residues were tested for cytotoxic potential on non-small lung cancer cell lines, A549 and H1299, and non-cancerous MRC5. DK-164 demonstrated remarkable selectivity toward cancer cells and more pronounced cytotoxicity against A549. The cytotoxicity of CC-78 toward H1299 was even higher than that of the well-established anticancer drugs cisplatin and tamoxifen, but it did not reveal any noticeable selective effect. DK-164 showed predominantly pro-apoptotic activity in non-small cell lung carcinoma (NSCLC) cells, while CC-78 caused accidental cell death with features characteristic of necrosis. The level of induced autophagy was similar for both substances in cancer cells. DK-164 treatment of A549, H1299, and MRC5 cells for 48 h significantly increased the fluorescence signal of the NFkB (nuclear factor ‘kappa-light-chain-enhancer’ of activated B-cells) protein in the nucleus in all three cell lines, while CC-78 did not provoke NFkB translocation in any of the tested cell lines. Both compounds caused a significant transfer of the p53 protein in the nucleus of A549 cells but not in non-cancerous MRC5 cells. In A549, DK-164 generated oxidative stress close to the positive control after 48 h, while CC-78 had a moderate effect on the cellular redox status. In the non-cancerous cells, MRC5, both compounds produced ROS similar to the positive control for the same incubation period. The different results related to the cytotoxic potential of DK-164 and CC-78 associated with the examined cellular mechanisms induced in lung cancer cells might be used to conclude the specific functions of the various functional groups in the ferrocene compounds, which can offer new perspectives for the design of antitumour drugs.
Collapse
Affiliation(s)
- Maria Schröder
- Institute of Molecular Biology “Akad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 21, 1113 Sofia, Bulgaria; (M.S.); (M.P.); (Z.V.); (E.P.)
| | - Maria Petrova
- Institute of Molecular Biology “Akad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 21, 1113 Sofia, Bulgaria; (M.S.); (M.P.); (Z.V.); (E.P.)
| | - Zlatina Vlahova
- Institute of Molecular Biology “Akad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 21, 1113 Sofia, Bulgaria; (M.S.); (M.P.); (Z.V.); (E.P.)
| | - Georgi M. Dobrikov
- Institute of Organic Chemistry with Center of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 9, 1113 Sofia, Bulgaria; (G.M.D.); (I.S.)
| | - Ivaylo Slavchev
- Institute of Organic Chemistry with Center of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 9, 1113 Sofia, Bulgaria; (G.M.D.); (I.S.)
| | - Evdokia Pasheva
- Institute of Molecular Biology “Akad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 21, 1113 Sofia, Bulgaria; (M.S.); (M.P.); (Z.V.); (E.P.)
| | - Iva Ugrinova
- Institute of Molecular Biology “Akad. Roumen Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str, bl 21, 1113 Sofia, Bulgaria; (M.S.); (M.P.); (Z.V.); (E.P.)
- Correspondence: ; Tel.: +359-887-985-463
| |
Collapse
|
13
|
Guan P, Zhou J, Girel S, Zhu X, Schwab M, Zhang K, Wang-Müller Q, Bigler L, Nick P. Anti-microtubule activity of the traditional Chinese medicine herb Northern Ban Lan (Isatis tinctoria) leads to glucobrassicin. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:2058-2074. [PMID: 34636476 DOI: 10.1111/jipb.13177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
Traditional Chinese medicine (TCM) belongs to the most elaborate and extensive systems of plant-based healing. The herb Northern Ban Lan (Isatis tinctoria) is famous for its antiviral and anti-inflammatory activity. Although numerous components isolated from I. tinctoria have been characterized so far, their modes of action have remained unclear. Here, we show that extracts from I. tinctoria exert anti-microtubular activity. Using time-lapse microscopy in living tobacco BY-2 (Nicotiana tabacum L. cv Bright Yellow 2) cells expressing green fluorescent protein-tubulin, we use activity-guided fractionation to screen out the biologically active compounds of I. tinctoria. Among 54 fractions obtained from either leaves or roots of I. tinctoria by methanol (MeOH/H2 O 8:2), or ethyl acetate extraction, one specific methanolic root fraction was selected, because it efficiently and rapidly eliminated microtubules. By combination of further purification with ultra-high-performance liquid chromatography and high-resolution tandem mass spectrometry most of the bioactivity could be assigned to the glucosinolate compound glucobrassicin. Glucobrassicin can also affect microtubules and induce apoptosis in HeLa cells. In the light of these findings, the antiviral activity of Northern Ban Lan is discussed in the context of microtubules being hijacked by many viral pathogens for cell-to-cell spread.
Collapse
Affiliation(s)
- Pingyin Guan
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Jianning Zhou
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Sergey Girel
- Department of Chemistry, University of Zürich, Winterthurerstr.190, CH-8057, Zürich, Switzerland
| | - Xin Zhu
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Marian Schwab
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Kunxi Zhang
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Qiyan Wang-Müller
- Research Institute of Organic Agriculture FiBL, Ackerstrasse 113, CH-5070, Frick, Switzerland
| | - Laurent Bigler
- Department of Chemistry, University of Zürich, Winterthurerstr.190, CH-8057, Zürich, Switzerland
| | - Peter Nick
- Molecular Cell Biology, Botanical Institute, Karlsruhe Institute of Technology, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| |
Collapse
|
14
|
Anti-Inflammatory Mechanisms of Novel Synthetic Ruthenium Compounds. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112110092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Inflammation is the primary biological reaction to induce severe infection or injury in the immune system. Control of different inflammatory cytokines, such as nitric oxide (NO), interleukins (IL), tumor necrosis factor alpha-(TNF-α), noncytokine mediator, prostaglandin E2 (PGE2), mitogen activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB), facilitates anti-inflammatory effect of different substances. Coordination metal complexes have been applied as metallo-drugs. Several metal complexes have found to possess potent biological activities, especially anticancer, cardioprotective, chondroprotective and anti-parasitosis activities. Among the metallo drugs, ruthenium-based (Ru) complexes have paid much attention in clinical applications. Despite the kinetic nature of Ru complexes is similar to platinum in terms of cell division events, their toxic effect is lower than that of cisplatin. This paper reviews the anti-inflammatory effect of novel synthetic Ru complexes with potential molecular mechanisms that are actively involved.
Collapse
|
15
|
Salameh J, Cantaloube I, Benoit B, Poüs C, Baillet A. Cdc42 and its BORG2 and BORG3 effectors control the subcellular localization of septins between actin stress fibers and microtubules. Curr Biol 2021; 31:4088-4103.e5. [PMID: 34329591 DOI: 10.1016/j.cub.2021.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/28/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023]
Abstract
Cell resistance to taxanes involves several complementary mechanisms, among which septin relocalization from actin stress fibers to microtubules plays an early role. By investigating the molecular mechanism underlying this relocalization, we found that acute paclitaxel treatment triggers the release from stress fibers and subsequent proteasome-mediated degradation of binder of Rho GTPases 2 (BORG2)/Cdc42 effector protein 3 (Cdc42EP3) and to a lesser extent of BORG3/Cdc42EP5, two Cdc42 effectors that link septins to actin in interphase cells. BORG2 or BORG3 silencing not only caused septin detachment from stress fibers but also mimicked the effects of paclitaxel by triggering both septin relocalization to microtubules and significant drug resistance. Conversely, BORG2 or BORG3 overexpression retained septins on actin fibers even after paclitaxel treatment, without affecting paclitaxel sensitivity. We found that drug-induced inhibition of Cdc42 resulted in a drop in BORG2 level and in the relocalization of septins to microtubules. Accordingly, although septins relocalized when overexpressing an inactive mutant of Cdc42, the expression of a constitutively active mutant acted locally at actin stress fibers to prevent septin release, even after paclitaxel treatment. These findings reveal the role of Cdc42 upstream of BORG2 and BORG3 in controlling the interplay between septins, actin fibers, and microtubules in basal condition and in response to taxanes.
Collapse
Affiliation(s)
- Joëlle Salameh
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Isabelle Cantaloube
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Béatrice Benoit
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Christian Poüs
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France; Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Clamart, France.
| | - Anita Baillet
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France.
| |
Collapse
|
16
|
Shokrollahi M, Mekhail K. Interphase microtubules in nuclear organization and genome maintenance. Trends Cell Biol 2021; 31:721-731. [PMID: 33902985 DOI: 10.1016/j.tcb.2021.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Microtubules are major cytoskeletal components mediating fundamental cellular processes, including cell division. Recent evidence suggests that microtubules also regulate the nucleus during the cell cycle's interphase stage. Deciphering such roles of microtubules should uncover direct crosstalk between the nucleus and cytoplasm, impacting genome function and organismal health. Here, we review emerging roles for microtubules in interphase genome regulation. We explore how microtubules exert cytoplasmic forces on the nucleus or transport molecular cargo, including DNA, into or within the nucleus. We also describe how microtubules perform these functions by establishing transient or stable connections with nuclear envelope elements. Lastly, we discuss how the regulation of the nucleus by microtubules impacts genome organization and repair. Together, the literature indicates that interphase microtubules are critical regulators of nuclear structure and genome stability.
Collapse
Affiliation(s)
- Mitra Shokrollahi
- Department of Laboratory Medicine and Pathobiology, MaRS Centre, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, MaRS Centre, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Canada Research Chairs Program, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
17
|
Mahendra CK, Tan LTH, Lee WL, Yap WH, Pusparajah P, Low LE, Tang SY, Chan KG, Lee LH, Goh BH. Angelicin-A Furocoumarin Compound With Vast Biological Potential. Front Pharmacol 2020; 11:366. [PMID: 32372949 PMCID: PMC7176996 DOI: 10.3389/fphar.2020.00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Angelicin, a member of the furocoumarin group, is related to psoralen which is well known for its effectiveness in phototherapy. The furocoumarins as a group have been studied since the 1950s but only recently has angelicin begun to come into its own as the subject of several biological studies. Angelicin has demonstrated anti-cancer properties against multiple cell lines, exerting effects via both the intrinsic and extrinsic apoptotic pathways, and also demonstrated an ability to inhibit tubulin polymerization to a higher degree than psoralen. Besides that, angelicin too demonstrated anti-inflammatory activity in inflammatory-related respiratory and neurodegenerative ailments via the activation of NF-κB pathway. Angelicin also showed pro-osteogenesis and pro-chondrogenic effects on osteoblasts and pre-chondrocytes respectively. The elevated expression of pro-osteogenic and chondrogenic markers and activation of TGF-β/BMP, Wnt/β-catenin pathway confirms the positive effect of angelicin bone remodeling. Angelicin also increased the expression of estrogen receptor alpha (ERα) in osteogenesis. Other bioactivities, such as anti-viral and erythroid differentiating properties of angelicin, were also reported by several researchers with the latter even displaying an even greater aptitude as compared to the commonly prescribed drug, hydroxyurea, which is currently on the market. Apart from that, recently, a new application for angelicin against periodontitis had been studied, where reduction of bone loss was indirectly caused by its anti-microbial properties. All in all, angelicin appears to be a promising compound for further studies especially on its mechanism and application in therapies for a multitude of common and debilitating ailments such as sickle cell anaemia, osteoporosis, cancer, and neurodegeneration. Future research on the drug delivery of angelicin in cancer, inflammation and erythroid differentiation models would aid in improving the bioproperties of angelicin and efficacy of delivery to the targeted site. More in-depth studies of angelicin on bone remodeling, the pro-osteogenic effect of angelicin in various bone disease models and the anti-viral implications of angelicin in periodontitis should be researched. Finally, studies on the binding of angelicin toward regulatory genes, transcription factors, and receptors can be done through experimental research supplemented with molecular docking and molecular dynamics simulation.
Collapse
Affiliation(s)
- Camille Keisha Mahendra
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Loh Teng Hern Tan
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Liang Ee Low
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Siah Ying Tang
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Subang Jaya, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Malaysia
| | - Kok Gan Chan
- International Genome Centre, Jiangsu University, Zhenjiang, China
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Learn Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
18
|
Hsia CH, Jayakumar T, Sheu JR, Hsia CW, Huang WC, Velusamy M, Lien LM. Synthetic Ruthenium Complex TQ-6 Potently Recovers Cerebral Ischemic Stroke: Attenuation of Microglia and Platelet Activation. J Clin Med 2020; 9:jcm9040996. [PMID: 32252398 PMCID: PMC7230480 DOI: 10.3390/jcm9040996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Activated microglia are crucial in the regulation of neuronal homeostasis and neuroinflammation. They also contribute to neuropathological processes after ischemic stroke. Thus, finding new approaches for reducing neuroinflammation has gained considerable attention. The metal ruthenium has gained notable attention because of its ability to form new complexes that can be used in disease treatment. [Ru(η6-cymene)2-(1H-benzoimidazol-2-yl)-quinoline Cl]BF4 (TQ-6), a potent ruthenium (II)-derived compound, was used in this study to investigate its neuroprotective action against microglia activation, middle cerebral artery occlusion (MCAO)-induced embolic stroke, and platelet activation, respectively. TQ-6 (2 μM) potently diminished inflammatory mediators (nitric oxide/inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2)) expression, nuclear factor kappa B (NF-κB) p65 phosphorylation, nuclear translocation, and hydroxyl radical (OH•) formation in LPS-stimulated microglia. Conversely, TQ-6 increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Moreover, it significantly reduced brain infarct volume and edema in MCAO mice. Additionally, it drastically inhibited platelet aggregation and OH• production in mice platelets. This study confirmed that TQ-6 exerts an anti-neuroinflammatory effect on microglia activation through neuroprotection, antiplatelet activation, and free radical scavenging. The authors propose that TQ-6 might mitigate neurodegenerative pathology by inhibiting the NF-κB-mediated downstream pathway (iNOS and COX-2) and enhancing Nrf2/HO-1 signaling molecules in microglia.
Collapse
Affiliation(s)
- Chih-Hsuan Hsia
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Chih-Wei Hsia
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Wei-Chieh Huang
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India;
| | - Li-Ming Lien
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
- Correspondence: ; Tel.: +886-2-283-322-11 (ext. 2071)
| |
Collapse
|
19
|
Hasanain M, Sahai R, Pandey P, Maheshwari M, Choyal K, Gandhi D, Singh A, Singh K, Mitra K, Datta D, Sarkar J. Microtubule disrupting agent-mediated inhibition of cancer cell growth is associated with blockade of autophagic flux and simultaneous induction of apoptosis. Cell Prolif 2020; 53:e12749. [PMID: 32167212 PMCID: PMC7162801 DOI: 10.1111/cpr.12749] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/14/2019] [Accepted: 11/29/2019] [Indexed: 12/16/2022] Open
Abstract
Objectives Given that autophagy inhibition is a feasible way to enhance sensitivity of cancer cells towards chemotherapeutic agents, identifying potent autophagy inhibitor has obvious clinical relevance. Here, we investigated ability of TN‐16, a microtubule disrupting agent, on modulation of autophagic flux and its significance in promoting in vitro and in vivo cancer cell death. Materials and methods The effect of TN‐16 on cancer cell proliferation, cell division, autophagic process and apoptotic signalling was assessed by various biochemical (Western blot and SRB assay), morphological (TEM, SEM, confocal microscopy) and flowcytometric assays. In vivo anti‐tumour efficacy of TN‐16 was investigated in syngeneic mouse model of breast cancer. Results TN‐16 inhibited cancer cell proliferation by impairing late‐stage autophagy and induction of apoptosis. Inhibition of autophagic flux was demonstrated by accumulation of autophagy‐specific substrate p62 and lack of additional LC3‐II turnover in the presence of lysosomotropic agent. The effect was validated by confocal micrographs showing diminished autophagosome‐lysosome fusion. Further studies revealed that TN‐16–mediated inhibition of autophagic flux promotes apoptotic cell death. Consistent with in vitro data, results of our in vivo study revealed that TN‐16–mediated tumour growth suppression is associated with blockade of autophagic flux and enhanced apoptosis. Conclusions Our data signify that TN‐16 is a potent autophagy flux inhibitor and might be suitable for (pre‐) clinical use as standard inhibitor of autophagy with anti‐cancer activity.
Collapse
Affiliation(s)
- Mohammad Hasanain
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Rohit Sahai
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Praveen Pandey
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Mayank Maheshwari
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Kuldeep Choyal
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Deepa Gandhi
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Akhilesh Singh
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Kavita Singh
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Kalyan Mitra
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India.,Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Dipak Datta
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Jayanta Sarkar
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
20
|
Lee SY, Kim S, Lim Y, Yoon HN, Ku NO. Keratins regulate Hsp70-mediated nuclear localization of p38 mitogen-activated protein kinase. J Cell Sci 2019; 132:jcs.229534. [PMID: 31427430 DOI: 10.1242/jcs.229534] [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: 01/03/2019] [Accepted: 08/12/2019] [Indexed: 12/31/2022] Open
Abstract
Intermediate filament protein keratin 8 (K8) binds to heat shock protein 70 (Hsp70) and p38 MAPK, and is phosphorylated at Ser74 by p38α (MAPK14, hereafter p38). However, a p38 binding site on K8 and the molecular mechanism of K8-p38 interaction related to Hsp70 are unknown. Here, we identify a p38 docking site on K8 (Arg148/149 and Leu159/161) that is highly conserved in other intermediate filaments. A docking-deficient K8 mutation caused increased p38-Hsp70 interaction and enhanced p38 nuclear localization, indicating that the p38 dissociated from mutant K8 makes a complex with Hsp70, which is known as a potential chaperone for p38 nuclear translocation. Comparison of p38 MAPK binding with keratin variants associated with liver disease showed that the K18 I150V variant dramatically reduced binding with p38, which is similar to the effect of the p38 docking-deficient mutation on K8. Because the p38 docking site on K8 (Arg148/149 and Leu159/161) and the K18 Ile150 residue are closely localized in the parallel K8/K18 heterodimer, the K18 I150V mutation might interfere with K8-p38 interaction. These findings show that keratins, functioning as cytoplasmic anchors for p38, modulate p38 nuclear localization and thereby might affect a number of p38-mediated signal transduction pathways.
Collapse
Affiliation(s)
- So-Young Lee
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Sujin Kim
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Younglan Lim
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Han-Na Yoon
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea .,Department of Bio-Convergence ISED, Underwood International College, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
21
|
Malacrida A, Meregalli C, Rodriguez-Menendez V, Nicolini G. Chemotherapy-Induced Peripheral Neuropathy and Changes in Cytoskeleton. Int J Mol Sci 2019; 20:E2287. [PMID: 31075828 PMCID: PMC6540147 DOI: 10.3390/ijms20092287] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/23/2022] Open
Abstract
Despite the different antineoplastic mechanisms of action, peripheral neurotoxicity induced by all chemotherapy drugs (anti-tubulin agents, platinum compounds, proteasome inhibitors, thalidomide) is associated with neuron morphological changes ascribable to cytoskeleton modifications. The "dying back" degeneration of distal terminals (sensory nerves) of dorsal root ganglia sensory neurons, observed in animal models, in in vitro cultures and biopsies of patients is the most evident hallmark of the perturbation of the cytoskeleton. On the other hand, in highly polarized cells like neurons, the cytoskeleton carries out its role not only in axons but also has a fundamental role in dendrite plasticity and in the organization of soma. In the literature, there are many studies focused on the antineoplastic-induced alteration of microtubule organization (and consequently, fast axonal transport defects) while very few studies have investigated the effect of the different classes of drugs on microfilaments, intermediate filaments and associated proteins. Therefore, in this review, we will focus on: (1) Highlighting the fundamental role of the crosstalk among the three filamentous subsystems and (2) investigating pivotal cytoskeleton-associated proteins.
Collapse
Affiliation(s)
- Alessio Malacrida
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Cristina Meregalli
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Virginia Rodriguez-Menendez
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| | - Gabriella Nicolini
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy.
| |
Collapse
|
22
|
Schröder M, Yusein-Myashkova S, Petrova M, Dobrikov G, Kamenova-Nacheva M, Todorova J, Pasheva E, Ugrinova I. The Effect of a Ferrocene Containing Camphor Sulfonamide DK-164 on Breast Cancer Cell Lines. Anticancer Agents Med Chem 2019; 19:1874-1886. [PMID: 31339077 DOI: 10.2174/1871520619666190724094334] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/25/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Drug resistance is a major cause of cancer treatment failure. Most cancer therapies involve multiple agents, to overcome it. Compounds that exhibit strong anti-tumor effect without damaging normal cells are more and more in the focus of research. Chemotherapeutic drugs, combining different moieties and functional groups in one molecule, can modulate different regulatory pathways in the cell and thus reach the higher efficacy than the agents, which affect only one cellular process. METHODS We tested the effect of recently synthesized ferrocene-containing camphor sulfonamide DK-164 on two breast cancer and one breast non-cancer cell lines. The cytotoxic effects were evaluated using the standard MTT-dye reduction and clonogenic assays. The apoptotic or autophagic effects were evaluated by Annexin v binding or LC3 puncta formation assays, respectively. Cell cycle arrest was determined using flow cytometry. Western blot and immunofluorescent analyses were used to estimate the localization and cellular distribution of key regulatory factors NFκB and p53. RESULTS Compound DK-164 has well pronounced cytotoxicity greater to cancer cells (MDA-MB-231 and MCF-7) compared to non-cancerous (MCF-10A). The IC50 value of the substance caused a cell cycle arrest in G1 phase and induced apoptosis up to 24 hours in both tumor cells, although being more pronounced in MCF-7, a functional p53 cell line. Treatment with IC50 concentration of the compound provoked autophagy in both tumor lines but is better pronounced in the more aggressive cancer line (MDA-MB-231). CONCLUSION The tested compound DK-164 showed promising properties as a potential therapeutic agent.
Collapse
Affiliation(s)
- Maria Schröder
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Shazie Yusein-Myashkova
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Maria Petrova
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Georgi Dobrikov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, bl. 9, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Mariana Kamenova-Nacheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, bl. 9, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Jordana Todorova
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Evdokia Pasheva
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| | - Iva Ugrinova
- Institute of Molecular Biology, "Roumen Tsanev'', Bulgarian Academy of Sciences, bl. 21, Acad. G. Bonchev Str., Sofia 1113, Bulgaria
| |
Collapse
|
23
|
Barvitenko N, Lawen A, Aslam M, Pantaleo A, Saldanha C, Skverchinskaya E, Regolini M, Tuszynski JA. Integration of intracellular signaling: Biological analogues of wires, processors and memories organized by a centrosome 3D reference system. Biosystems 2018; 173:191-206. [PMID: 30142359 DOI: 10.1016/j.biosystems.2018.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/03/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Myriads of signaling pathways in a single cell function to achieve the highest spatio-temporal integration. Data are accumulating on the role of electromechanical soliton-like waves in signal transduction processes. Theoretical studies strongly suggest feasibility of both classical and quantum computing involving microtubules. AIM A theoretical study of the role of the complex composed of the plasma membrane and the microtubule-based cytoskeleton as a system that transmits, stores and processes information. METHODS Theoretical analysis presented here refers to (i) the Penrose-Hameroff theory of consciousness (Orchestrated Objective Reduction; Orch OR), (ii) the description of the centrosome as a reference system for construction of the 3D map of the cell proposed by Regolini, (iii) the Heimburg-Jackson model of the nerve pulse propagation along axons' lipid bilayer as soliton-like electro-mechanical waves. RESULTS AND CONCLUSION The ideas presented in this paper provide a qualitative model for the decision-making processes in a living cell undergoing a differentiation process. OUTLOOK This paper paves the way for the real-time live-cell observation of information processing by microtubule-based cytoskeleton and cell fate decision making.
Collapse
Affiliation(s)
| | - Alfons Lawen
- Monash University, School of Biomedical Sciences, Department of Biochemistry and Molecular Biology, VIC, 3800, Australia
| | - Muhammad Aslam
- Medical Clininc I, Cardiology/Angiology, University Hospital, Justus-Liebig-University, Giessen, Germany
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Carlota Saldanha
- Instituto de Medicina Molecular, Instituto de Bioquimica, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Marco Regolini
- Department of Bioengineering and Mathematical Modeling, AudioLogic, Milan, Italy
| | - Jack A Tuszynski
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada; Department of Physics, University of Alberta, Edmonton, Alberta, Canada; Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, IT-10128, Torino, Italy.
| |
Collapse
|
24
|
El-Kashef DH. Nicorandil alleviates ovalbumin-induced airway inflammation in a mouse model of asthma. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 59:132-137. [PMID: 29579542 DOI: 10.1016/j.etap.2018.03.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/18/2018] [Indexed: 06/08/2023]
Abstract
Nicorandil is an antianginal drug that has anti-inflammatory property. This study aimed to investigate the effects of nicorandil on allergic asthma induced by ovalbumin (OVA) in mice in comparison with dexamethasone. Mice were sensitized to OVA (on days 0 and 7) and challenged with OVA three times (on days 14, 15 and 16). Nicorandil was given orally for 5 days 1 h before OVA treatment in days of challenge. Progression of asthma was accompanied by significant elevation in the lung/body weight index, LDH, total protein, IL-13 and NF-κB levels besides inflammatory cell counts in BALF; Also pulmonary MDA and NO contents were significantly increased but GSH and SOD levels were decreased. Histopathological alterations in lung tissues were also observed. In contrast, nicorandil treatment significantly alleviated OVA-induced lung injury. In conclusion, our results proposed that nicorandil is equivalent to dexamethasone in ameliorating allergic asthma by restoring oxidant/antioxidant balance and reducing inflammation.
Collapse
Affiliation(s)
- Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
25
|
Gilmore SP, Gonye ALK, Li EC, Espinosa de Los Reyes S, Gupton JT, Quintero OA, Fischer-Stenger K. Effects of a novel microtubule-depolymerizer on pro-inflammatory signaling in RAW264.7 macrophages. Chem Biol Interact 2018; 280:109-116. [PMID: 29247640 PMCID: PMC5766364 DOI: 10.1016/j.cbi.2017.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/04/2017] [Accepted: 12/12/2017] [Indexed: 12/21/2022]
Abstract
The Nuclear Factor-kappa B (NF-κB) pathway is vital for immune system regulation and pro-inflammatory signaling. Many inflammatory disorders and diseases, including cancer, are linked to dysregulation of NF-κB signaling. When macrophages recognize the presence of a pathogen, the signaling pathway is activated, resulting in the nuclear translocation of the transcription factor, NF-κB, to turn on pro-inflammatory genes. Here, we demonstrate the effects of a novel microtubule depolymerizer, NT-07-16, a polysubstituted pyrrole compound, on this process. Treatment with NT-07-16 decreased the production of pro-inflammatory cytokines in RAW264.7 mouse macrophages. It appears that the reduction in pro-inflammatory mediators produced by the macrophages after exposure to NT-07-16 may be due to activities upstream of the translocation of NF-κB into the nucleus. NF-κB translocation occurs after its inhibitory protein, IκB-α is phosphorylated which signals for its degradation releasing NF-κB so it is free to move into the nucleus. Previous studies from other laboratories indicate that these processes are associated with the microtubule network. Our results show that exposure to the microtubule-depolymerizer, NT-07-16 reduces the phosphorylation of IκB-α and also decreases the association of NF-κB with tubulin which may affect the ability of NF-κB to translocate into the nucleus. Therefore, the anti-inflammatory activity of NT-07-16 may be explained, at least in part, by alterations in these steps in the NF-κB signaling pathway leading to less NF-κB entering the nucleus and reducing the production of pro-inflammatory mediators by the activated macrophages.
Collapse
Affiliation(s)
| | - Anna L K Gonye
- Department of Biology, University of Richmond, VA 23173, USA.
| | - Elizabeth C Li
- Department of Biology, University of Richmond, VA 23173, USA.
| | | | - John T Gupton
- Department of Chemistry, University of Richmond, VA 23173, USA.
| | - Omar A Quintero
- Department of Biology, University of Richmond, VA 23173, USA.
| | | |
Collapse
|
26
|
He X, Gong P, Wei Z, Liu W, Wang W, Li J, Yang Z, Zhang X. Peroxisome proliferator-activated receptor-γ-mediated polarization of macrophages in Neospora caninum infection. Exp Parasitol 2017; 178:37-44. [PMID: 28527905 DOI: 10.1016/j.exppara.2017.05.002] [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: 10/25/2016] [Revised: 04/25/2017] [Accepted: 05/16/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Neospora caninum is an apicomplexan parasite closely related Toxoplasma gondii, which causes neurological disease and abortion in multiple animal species. Macrophage polarization plays an important role in host immune responses to parasites infection, such as Toxoplasma gondii, Leishmania, Trypanosoma cruzi. However, the dynamics of macrophage polarization, as well as the possible mechanism that regulate macrophage polarization, during N. caninum infection remains unclear. METHODS The M1 and M2-phenotypic markers of peritoneal macrophages from mice infected with tachyzoites of Nc-1 were analyzed by flow cytometry (FCM) analysis. Then J774A.1 cells were respectively treated with GW9662 and RGZ, and stimulated by tachyzoites of Nc-1. M1 and M2-phenotypic markers were determined by FCM and ELISA. And the activations of PPAR-γ and NF-κB were determined by Western blotting. RESULTS In this study, our data showed that macrophages were preferentially differentiated into the M1 type during the acute stage of N. caninum infection, while the level of M2 macrophages significantly increased during the chronic stage of infection. In vitro study, compared with the GW9662 group and RGZ group, N. caninum can promote M2-polarized phenotype through up-regulate the activity of PPAR-γ and inhibting NF-κB activation. CONCLUSION In conclusion, this study demonstrated that macrophages are plastic since M1 differentiated macrophages can express M2 markers with N. caninum infection through up-regulating the activity of PPAR-γ and inhibting NF-κB activation and may be providing new insights for the prevention and treatment of N. caninum infection.
Collapse
Affiliation(s)
- Xuexiu He
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Pengtao Gong
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhengkai Wei
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Weijian Liu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Weili Wang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jianhua Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhengtao Yang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
27
|
Wei DZ, Guo XY, Lin LN, Lin MX, Gong YQ, Ying BY, Huang MY. Effects of Angelicin on Ovalbumin (OVA)-Induced Airway Inflammation in a Mouse Model of Asthma. Inflammation 2017; 39:1876-1882. [PMID: 27581277 DOI: 10.1007/s10753-016-0423-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Angelicin, a furocoumarin found in Psoralea corylifolia L. fruit, has been reported to have anti-inflammatory activity. The purpose of this study was to determine the protective effects of angelicin on allergic asthma induced by ovalbumin (OVA) in mice. Mice were sensitized to OVA (on days 0 and 14) and challenged with OVA three times (on days 21 to 23). Angelicin (2.5, 5, 10 mg/kg) was given intraperitoneally 1 h before OVA treatment after the initial OVA sensitization. The production of IL-4, IL-5, and IL-13 in BALF and IgE in the serum were measured by ELISA. Lung histological changes were detected by using hematoxylin and eosin (H&E) stain. The results showed that angelicin significantly inhibited inflammatory cells infiltration into the lungs. Histological studies showed that angelicin significantly attenuated OVA-induced lung injury. Meanwhile, treatment of angelicin dose-dependently inhibited OVA-induced the production of IL-4, IL-5, and IL-13 in BALF and IgE in the serum. Furthermore, angelicin was found to inhibit airway hyperresponsiveness and NF-kB activation. In conclusion, our results suggested that angelicin inhibited allergic airway inflammation and hyperresponsiveness by inhibiting NF-kB activation.
Collapse
Affiliation(s)
- Da-Zhen Wei
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xian-Yang Guo
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Li-Na Lin
- Department of Neurology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Meng-Xiang Lin
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yu-Qiang Gong
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Bin-Yu Ying
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ming-Yuan Huang
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
28
|
C1, a highly potent novel curcumin derivative, binds to tubulin, disrupts microtubule network and induces apoptosis. Biosci Rep 2016; 36:BSR20160039. [PMID: 26980197 PMCID: PMC4847174 DOI: 10.1042/bsr20160039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/15/2016] [Indexed: 12/15/2022] Open
Abstract
C1 is one of the most potent curcumin analogues identified till date which inhibits proliferation of various cancer cell lines. C1 binds to tubulin and depolymerized microtubules of MCF-7 cells. C1 altered the expression of apoptotic proteins and induces p53-dependent apoptosis. We have synthesized a curcumin derivative, 4-{5-(4-hydroxy-3-methoxy-phenyl)-2-[3-(4-hydroxy-3-methoxy-phenyl)-acryloyl]-3-oxo-penta-1,4-dienyl}-piperidine-1-carboxylic acid tert-butyl ester (C1) that displays much stronger antiproliferative activity against various types of cancer cells including multidrug resistance cells than curcumin. C1 depolymerized both interphase and mitotic microtubules in MCF-7 cells and also inhibited the reassembly of microtubules in these cells. C1 inhibited the polymerization of purified tubulin, disrupted the lattice structure of microtubules and suppressed their GTPase activity in vitro. The compound bound to tubulin with a dissociation constant of 2.8±1 μM and perturbed the secondary structures of tubulin. Further, C1 treatment reduced the expression of Bcl2, increased the expression of Bax and down regulated the level of a key regulator of p53, murine double minute 2 (Mdm2) (S166), in MCF-7 cells. C1 appeared to induce p53 mediated apoptosis in MCF-7 cells. Interestingly, C1 showed more stability in aqueous buffer than curcumin. The results together showed that C1 perturbed microtubule network and inhibited cancer cells proliferation more efficiently than curcumin. The strong antiproliferative activity and improved stability of C1 indicated that the compound may have a potential as an anticancer agent.
Collapse
|
29
|
Pacios S, Xiao W, Mattos M, Lim J, Tarapore RS, Alsadun S, Yu B, Wang CY, Graves DT. Osteoblast Lineage Cells Play an Essential Role in Periodontal Bone Loss Through Activation of Nuclear Factor-Kappa B. Sci Rep 2015; 5:16694. [PMID: 26666569 PMCID: PMC4678879 DOI: 10.1038/srep16694] [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: 09/22/2014] [Accepted: 09/09/2015] [Indexed: 11/25/2022] Open
Abstract
Bacterial pathogens stimulate periodontitis, the most common osteolytic disease in humans and the most common cause of tooth loss in adults. Previous studies identified leukocytes and their products as key factors in this process. We demonstrate for the first time that osteoblast lineage cells play a critical role in periodontal disease. Oral infection stimulated nuclear localization of NF-κB in osteoblasts and osteocytes in the periodontium of wild type but not transgenic mice that expressed a lineage specific dominant negative mutant of IKK (IKK-DN) in osteoblast lineage cells. Wild-type mice were also susceptible to bacteria induced periodontal bone loss but transgenic mice were not. The lack of bone loss in the experimental group was linked to reduced RANKL expression by osteoblast lineage cells that led to diminished osteoclast mediated bone resorption and greater coupled new bone formation. The results demonstrate that osteoblast lineage cells are key contributors to periodontal bone loss through an NF-κB mediated mechanism.
Collapse
Affiliation(s)
- Sandra Pacios
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Wenmei Xiao
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Periodontology, School and Hospital of Stomatology, Peking University, Beijing, China
| | - Marcelo Mattos
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jason Lim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rohinton S Tarapore
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarah Alsadun
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bo Yu
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
30
|
Shao G, Tian Y, Wang H, Liu F, Xie G. Protective effects of melatonin on lipopolysaccharide-induced mastitis in mice. Int Immunopharmacol 2015; 29:263-268. [PMID: 26590117 DOI: 10.1016/j.intimp.2015.11.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 10/18/2015] [Accepted: 11/06/2015] [Indexed: 12/17/2022]
Abstract
Melatonin, a secretory product of the pineal gland, has been reported to have antioxidant and anti-inflammatory effects. However, the protective effects of melatonin on lipopolysaccharide (LPS)-induced mastitis have not been reported. The purpose of this study was to investigate the anti-inflammatory effects and the underlying mechanisms of melatonin on LPS-induced mastitis both in vivo and in vitro. In vivo, our results showed that melatonin attenuated LPS-induced mammary histopathologic changes and myeloperoxidase (MPO) activity. Melatonin also inhibited LPS-induced inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) production in mammary tissues. In vitro, melatonin was found to inhibit LPS-induced TNF-α and IL-6 production in mouse mammary epithelial cells. Melatonin also suppressed LPS-induced Toll-like receptor 4 (TLR4) expression and nuclear factor-kappaB (NF-κB) activation in a dose-dependent manner. In addition, melatonin was found to up-regulate the expression of PPAR-γ. Inhibition of PPAR-γ by GW9662 reduced the anti-inflammatory effects of melatonin. In conclusion, we found that melatonin, for the first time, had protective effects on LPS-induced mastitis in mice. The anti-inflammatory mechanism of melatonin was through activating PPAR-γ which subsequently inhibited LPS-induced inflammatory responses.
Collapse
Affiliation(s)
- Guoxi Shao
- The Second Hospital of Jilin University, China
| | - Yinggang Tian
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China
| | - Haiyu Wang
- College of Veterinary Medicine, Jilin University, China
| | - Fangning Liu
- College of Veterinary Medicine, Jilin University, China
| | - Guanghong Xie
- College of Veterinary Medicine, Jilin University, China.
| |
Collapse
|
31
|
Zhang H, Yan J, Zhuang Y, Han G. Anti-inflammatory effects of farrerol on IL-1β-stimulated human osteoarthritis chondrocytes. Eur J Pharmacol 2015; 764:443-447. [DOI: 10.1016/j.ejphar.2015.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022]
|
32
|
He X, Wei Z, Zhou E, Chen L, Kou J, Wang J, Yang Z. Baicalein attenuates inflammatory responses by suppressing TLR4 mediated NF-κB and MAPK signaling pathways in LPS-induced mastitis in mice. Int Immunopharmacol 2015. [PMID: 26202808 DOI: 10.1016/j.intimp.2015.07.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Baicalein is a phenolic flavonoid presented in the dry roots of Scutellaria baicalensis Georgi. It has been reported that baicalein possesses a number of biological properties, such as antiviral, antioxidative, anti-inflammatory, antithrombotic, and anticancer properties. However, the effect of baicalein on mastitis has not yet been reported. This research aims to detect the effect of baicalein on lipopolysaccharide (LPS)-induced mastitis in mice and to investigate the molecular mechanisms. Baicalein was administered intraperitoneally 1h before and 12h after LPS treatment. The results indicated that baicalein treatment markedly attenuated the damage of the mammary gland induced by LPS, suppressed the activity of myeloperoxidase (MPO) and the levels of tumor necrosis factor (TNF-α) and interleukin (IL-1β) in mice with LPS-induced mastitis. Besides, baicalein blocked the expression of Toll-like receptor 4 (TLR4) and then suppressed the phosphorylation of nuclear transcription factor-kappaB (NF-κB) p65 and degradation inhibitor of NF-κBα (IκBα) and, and inhibited the phosphorylation of p38, extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) in mitogen-activated protein kinase (MAPK) signal pathway. These findings suggested that baicalein may have a potential prospect against mastitis.
Collapse
Affiliation(s)
- Xuexiu He
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Zhengkai Wei
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Ershun Zhou
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Libin Chen
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Jinhua Kou
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Jingjing Wang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Zhengtao Yang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China.
| |
Collapse
|
33
|
Linalool Inhibits LPS-Induced Inflammation in BV2 Microglia Cells by Activating Nrf2. Neurochem Res 2015; 40:1520-5. [DOI: 10.1007/s11064-015-1629-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/17/2015] [Accepted: 05/28/2015] [Indexed: 12/20/2022]
|