1
|
Wan TCR, Wei L, Cheng LH, Chin WC, Shen J, Chan FF, Kuang Z, Wang C, Wong CCL, Wong CM. Genome-wide CRISPR Screening Identifies NFκB and c-MET as Druggable Targets to Sensitize Lenvatinib Treatment in Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2025; 19:101502. [PMID: 40120675 DOI: 10.1016/j.jcmgh.2025.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC), the dominant form of liver cancer, is a leading cause of cancer death worldwide. Sorafenib and lenvatinib have long been the 2 limited options of first-line treatments for patients with unresectable advanced HCC. However, the single-drug treatment strategy only shows modest survival benefit, mostly because of the survival ability of cancer cells to activate alternative pathways for compensation. In this study, we aim to identify druggable targets contributing to lenvatinib resistance and evaluate the efficacy of combining respective inhibitors and lenvatinib on HCC. METHODS Genome-scale clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 knockout library screening was applied on the vehicle group and lenvatinib treatment group. Identified druggable candidates were validated individually on HCC cell models. Therapeutic effects of the combined treatment of inhibitors of candidate genes and lenvatinib were evaluated in vitro and in vivo. RESULTS We successfully identified NFKB1 and MET as critical drivers for the development of lenvatinib resistance in HCC cells. By perturbing the 2 genes with either CRISPR knockout or RNA interference approaches, lenvatinib treatments were significantly sensitized. Moreover, using small molecules QNZ and cabozantinib to target NFKB1 and MET, respectively, this together with lenvatinib could synergistically induce apoptosis and suppress HCC growth in vitro and in vivo. CONCLUSION Our results demonstrated that genome-wide CRISPR/Cas9 screening is a powerful tool for the design of rational combinational cancer therapy and provided candidate genes possible for combined treatments with lenvatinib to improve therapy efficacy.
Collapse
Affiliation(s)
- Ting-Chi Rebecca Wan
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Lai Wei
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Lai-Hung Cheng
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Wai-Ching Chin
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Jialing Shen
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - For-Fan Chan
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Zhijian Kuang
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Carmen Chak-Lui Wong
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Chun-Ming Wong
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Morgan I, Rennert R, Berger R, Jelača S, Maksimović-Ivanić D, Dunđerović D, Mijatović S, Kaluđerović GN, Wessjohann LA. The impact of 9-azaglycophymine and phenylguanidine derivatives on the proliferation of various breast cancer cell lines in vitro and in vivo. Sci Rep 2024; 14:28126. [PMID: 39548116 PMCID: PMC11568214 DOI: 10.1038/s41598-024-71624-8] [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: 04/23/2024] [Accepted: 08/29/2024] [Indexed: 11/17/2024] Open
Abstract
Quinazolinones, particularly 9-azaglycophymines, and closely related derivatives and precursors were tested in vitro against various breast cancer cell lines representing the major types of breast tumors. Among the 49 compounds tested, azaglycophymine derivative 19 with an electron-withdrawing substituent demonstrated the most significant anti-proliferative effects, with IC50 values of around 4 µM. Extensive cell-based investigations revealed that compound 19 induced caspase-dependent apoptosis in HCC1937 (human TNBC), BT-474 (human HER2+/HR+), and 4T1 (mouse TNBC) cells. In contrast, in MDA-MB-468 (human TNBC) and MCF-7 (human HR+) cells, the cell death was induced via a non-apoptotic pathway. The in vivo efficacy of compound 19 was validated using a syngeneic orthotopic 4T1 model in BALB/c mice, resulting in significant reduction of 4T1 breast tumor growth upon intraperitoneal (i.p.) application of doses of 5 or 20 mg/kg. These findings highlight the potential of compound 19 as a promising scaffold for the development of new therapeutic agents for various types of breast cancer and a first structure-activity insight.
Collapse
Affiliation(s)
- Ibrahim Morgan
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle (Saale), Germany
| | - Robert Rennert
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle (Saale), Germany.
| | - Robert Berger
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle (Saale), Germany
- , Berlin, Germany
| | - Sanja Jelača
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia (IBISS), University of Belgrade, Bulevar Despota Stefana 142, 11108, Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia (IBISS), University of Belgrade, Bulevar Despota Stefana 142, 11108, Belgrade, Serbia
| | - Duško Dunđerović
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000, Belgrade, Serbia
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia (IBISS), University of Belgrade, Bulevar Despota Stefana 142, 11108, Belgrade, Serbia
| | - Goran N Kaluđerović
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle (Saale), Germany
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Straße 2, 06217, Merseburg, Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120, Halle (Saale), Germany.
| |
Collapse
|
3
|
Zhao Y, Lv H, Yu C, Liang J, Yu H, Du Z, Zhang R. Systemic inhibition of mitochondrial fatty acid β-oxidation impedes zebrafish ventricle regeneration. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167442. [PMID: 39059593 DOI: 10.1016/j.bbadis.2024.167442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Unlike humans and other mammals, zebrafish demonstrate a remarkable capacity to regenerate their injured hearts throughout life. Mitochondrial fatty acid β-oxidation (FAO) contributes to major energy demands of the adult hearts under physiological conditions; however, its functions in regulating cardiac regeneration and the underlying mechanisms are not completely understood. Different strategies targeting FAO have yield mixed outcomes. Here, we demonstrated that pharmacological inhibition of mitochondrial FAO with mildronate (MD) caused lipid accumulation in zebrafish larvae and suppressed ventricle regeneration. MD treatment impeded cardiogenic factor reactivation and cardiomyocyte (CM) proliferation, and impaired ventricle regeneration could be rescued by exogenous l-carnitine supplementation. Moreover, compared with the ablated hearts of wild-type fish, ventricle regeneration, cardiogenic factor reactivation and CM proliferation were significantly blocked in the ablated hearts of carnitine palmitoyltransferase-1b (cpt1b) knockout zebrafish. Further experiments suggested that NF-κB signaling and increased inflammation may be involved in the impediment of ventricle regeneration caused by systemic mitochondrial FAO inhibition. Overall, our study demonstrates the essential roles of mitochondrial FAO in zebrafish ventricle regeneration and reaffirms the sophisticated and multifaceted roles of FAO in heart regeneration with regard to different injury models and means of FAO inhibition.
Collapse
Affiliation(s)
- Yan Zhao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hongbo Lv
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Chunxiao Yu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Jieling Liang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hong Yu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Zhenyu Du
- School of Life Sciences, East China Normal University, Shanghai, China.
| | - Ruilin Zhang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
4
|
Xu B, Anderson BM, Mintern JD, Edgington-Mitchell LE. TLR9-dependent dendritic cell maturation promotes IL-6-mediated upregulation of cathepsin X. Immunol Cell Biol 2024; 102:787-800. [PMID: 38979698 DOI: 10.1111/imcb.12806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Cysteine cathepsins are lysosomal proteases subject to dynamic regulation within antigen-presenting cells during the immune response and associated diseases. To investigate the regulation of cathepsin X, a carboxy-mono-exopeptidase, during maturation of dendritic cells (DCs), we exposed immortalized mouse DCs to various Toll-like receptor agonists. Using a cathepsin X-selective activity-based probe, sCy5-Nle-SY, we observed a significant increase in cathepsin X activation upon TLR-9 agonism with CpG, and to a lesser extent with Pam3 (TLR1/2), FSL-1 (TLR2/6) and LPS (TLR4). Despite clear maturation of DCs in response to Poly I:C (TLR3), cathepsin X activity was only slightly increased by this agonist, suggesting differential regulation of cathepsin X downstream of TLR activation. We demonstrated that cathepsin X was upregulated at the transcriptional level in response to CpG. This occurred at late time points and was not dampened by NF-κB inhibition. Factors secreted from CpG-treated cells were able to provoke cathepsin X upregulation when applied to naïve cells. Among these factors was IL-6, which on its own was sufficient to induce transcriptional upregulation and activation of cathepsin X. IL-6 is highly secreted by DCs in response to CpG but much less so in response to poly I:C, and inhibition of the IL-6 receptor subunit glycoprotein 130 prevented CpG-mediated cathepsin X upregulation. Collectively, these results demonstrate that cathepsin X is differentially transcribed during DC maturation in response to diverse stimuli, and that secreted IL-6 is critical for its dynamic regulation.
Collapse
Affiliation(s)
- Bangyan Xu
- Department of Biochemistry & Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Bethany M Anderson
- Department of Biochemistry & Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Justine D Mintern
- Department of Biochemistry & Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Laura E Edgington-Mitchell
- Department of Biochemistry & Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
5
|
El-Araby RE, Wasif K, Johnson R, Tu Q, Aboushousha T, Zhu ZX, Chen J. Establishment of a novel cellular model for Alzheimer's disease in vitro studies. Exp Neurol 2024; 378:114820. [PMID: 38789025 DOI: 10.1016/j.expneurol.2024.114820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/07/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory loss, cognitive impairment, and behavioral and psychological symptoms of dementia. The limited efficacy of drugs for the treatment of neurodegenerative diseases reflects their complex etiology and pathogenesis. A novel in vitro model may help to bridge the gap between existing preclinical animal models and human clinical trials, thus identifying promising therapeutic targets that can be explored in upcoming clinical trials. By assisting in the identification of the mechanism of action and potential dangers, in vitro testing can also shorten the time and expense of translation. AIM As a result of these factors, our objective is to develop a powerful and informative cellular model of AD within a short period of time. Through triggering the MAPK and NF-κβ signaling pathways with the aid of small chemical compounds (PAF C-16 and BetA), respectively, in mouse microglial (SIM-A9) and neuroblast Neuro-2a (N2a) cell lines. RESULTS PAF C-16, initiated an activation effect at a concentration of 3.12 nM to 25 nM in the SIM-A9 and N2a cell lines after 72 h. BetA, activated the NF-κβ pathway with a concentration of 12.5 nM to 25 nM in the SIM-A9 and N2a cell lines after 72 h. The combination of the activator chemicals provided suitable activation for MEK1/2-ERK and NF-κβ in more than three subcultures. Activators significantly initiate APP and MAPT gene expression, as well as the expression of proteins APP, β. Amyloid, tau, and p-tau. The activation of the targeted pathways leads to significant morphological changes. CONCLUSION We can infer that the MEK1/2-ERK and NF-κβ pathways, respectively, are directly activated by the PAF C-16 and BetA chemicals. The activation of MEK1/2-ERK pathway results in the activation of the APP gene, which in turn activates the β. Amyloid protein, which in turn results in plaque. Furthermore, NF-κβ activation results in the activation of the MAPT gene, which leads to Tau and p-Tau protein activation, which ultimately results in tangles. This can be put into practice in just three days, with a high level of activity and stability that is passed down to the next three generations (subculture), with significant morphological changes. In microglial and neuroblast cell lines, we were successful in creating a novel AD-cell model.
Collapse
Affiliation(s)
- Rady E El-Araby
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Komal Wasif
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Human Physiology, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA 02215, USA
| | - Rebecca Johnson
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Human Physiology, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA 02215, USA
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Tarek Aboushousha
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Zoe Xiaofang Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Genetics, Molecular and Cell Biology, Tufts University School of Medicine, and Graduate School of Biomedical Sciences. 136 Harrison Ave, M&V 830, Boston, MA 02111, USA.
| |
Collapse
|
6
|
Sprague JL, Schille TB, Allert S, Trümper V, Lier A, Großmann P, Priest EL, Tsavou A, Panagiotou G, Naglik JR, Wilson D, Schäuble S, Kasper L, Hube B. Candida albicans translocation through the intestinal epithelial barrier is promoted by fungal zinc acquisition and limited by NFκB-mediated barrier protection. PLoS Pathog 2024; 20:e1012031. [PMID: 38427950 PMCID: PMC10907035 DOI: 10.1371/journal.ppat.1012031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024] Open
Abstract
The opportunistic fungal pathogen Candida albicans thrives on human mucosal surfaces as a harmless commensal, but frequently causes infections under certain predisposing conditions. Translocation across the intestinal barrier into the bloodstream by intestine-colonizing C. albicans cells serves as the main source of disseminated candidiasis. However, the host and microbial mechanisms behind this process remain unclear. In this study we identified fungal and host factors specifically involved in infection of intestinal epithelial cells (IECs) using dual-RNA sequencing. Our data suggest that host-cell damage mediated by the peptide toxin candidalysin-encoding gene ECE1 facilitates fungal zinc acquisition. This in turn is crucial for the full virulence potential of C. albicans during infection. IECs in turn exhibit a filamentation- and damage-specific response to C. albicans infection, including NFκB, MAPK, and TNF signaling. NFκB activation by IECs limits candidalysin-mediated host-cell damage and mediates maintenance of the intestinal barrier and cell-cell junctions to further restrict fungal translocation. This is the first study to show that candidalysin-mediated damage is necessary for C. albicans nutrient acquisition during infection and to explain how IECs counteract damage and limit fungal translocation via NFκB-mediated maintenance of the intestinal barrier.
Collapse
Affiliation(s)
- Jakob L. Sprague
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Tim B. Schille
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Verena Trümper
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Adrian Lier
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Peter Großmann
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Antzela Tsavou
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Gianni Panagiotou
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Duncan Wilson
- Medical Research Council, Centre for Medical Mycology at the University of Exeter, Exeter, United Kingdom
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Hans-Knöll-Institute, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
7
|
Tang Y, Du E, Wang G, Qin F, Meng Z, Dai L, Wang Y, Ren S. A negative feedback loop centered on SMAD3 expression in transforming growth factor β1-induced corneal myofibroblast differentiation. Exp Eye Res 2023; 236:109654. [PMID: 37734427 DOI: 10.1016/j.exer.2023.109654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023]
Abstract
SMAD3 downregulation is documented in transforming growth factor β1 (TGF-β1)-induced corneal fibroblasts differentiation to myofibroblasts ("fibroTOmyoDiff") or corneal wound healing. However, the exact regulatory mechanism of TGF-β1/SMAD3 pathway in this context remains unclear. Here, we investigated the role and related mechanism of SMAD3 down-regulation in TGF-β1-induced human corneal fibroTOmyoDiff. By detecting expression changes of SMAD family during this process, we demonstrated that SMAD3 protein expression was dramatically decreased in the process and the decrease occurred mainly in SMAD3 gene transcription. Furthermore, SMAD3 overexpression using lentivirus infection and knockdown using sgRNA lentivirus infection or siRNAs revealed that SMAD3 overexpression enhanced TGF-β1-induced corneal fibroTOmyoDiff and vice versa. In addition, specific siRNAs and inhibitors targeting particular signaling pathway were used to figure out the intracellular signaling pathway regulating SMAD3, and the result showed that the decease of SMAD3 induced by TGF-β1 stimulation in human corneal fibroblasts (HCFs) was strikingly prevented by SMAD4 knockdown or p38 signaling inhibitor SB203580 treatment. Collectively, these results demonstrate that, in TGF-β1 induced corneal fibroTOmyoDiff, down-regulation of SMAD3 expression regulated by SMAD4 and p38 signaling pathways forms a negative feedback loop of TGFβ signaling to avoid excessive activation of the signaling, which suggest that SMAD3 may be a key target for corneal fibrosis treatment.
Collapse
Affiliation(s)
- Yunlan Tang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Enming Du
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Gang Wang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Fangyuan Qin
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhihong Meng
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lijuan Dai
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yiqiang Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| | - Shengwei Ren
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
8
|
Kerdphon S, Khamto N, Buddhachat K, Ngoenkam J, Paensuwan P, Pongcharoen S, Singh T, Meepowpan P, Jongcharoenkamol J. Structure-Activity Relationship and Molecular Docking of Quinazolinones Inhibiting Expression of COX-2, IL-1β, iNOS, and TNF-α through NF-κB Pathways. ACS Med Chem Lett 2023; 14:1167-1173. [PMID: 37736179 PMCID: PMC10510524 DOI: 10.1021/acsmedchemlett.3c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/10/2023] [Indexed: 09/23/2023] Open
Abstract
The quinazolinone scaffold is found in natural products and biologically active compounds, including inflammatory inhibitors. Major proteins or enzymes involved in the inflammation process are regulated by the amount of gene expression. Quinazolinone derivatives were investigated and developed against the inflammatory genes cyclooxygenase-2 (COX-2), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS) in the lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cell line. The mRNA expressions were measured using a real-time quantitative polymerase chain reaction (RT-qPCR). Quinazolinone compounds at 62.5 μM demonstrated anti-COX-2 and anti-IL-1β mRNA expressions down to 0.50% and 3.10% gene expression, respectively, via inhibition of nuclear factor κB (NF-κB). Molecular docking was performed to explain the interaction between the binding site and the developed compounds as well as the structure-activity relationship of the quinazolinone moiety.
Collapse
Affiliation(s)
- Sutthichat Kerdphon
- Department
of Chemistry, Faculty of Science, Naresuan
University, Phitsanulok 65000, Thailand
- Center
of Excellence in Petroleum, Petrochemicals and Advanced Materials,
Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Nopawit Khamto
- Department
of Chemistry, Faculty of Science, Chiang
Mai University, Chiang
Mai 50200, Thailand
- Graduate
School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kittisak Buddhachat
- Department
of Biology, Faculty of Science, Naresuan
University, Phitsanulok 65000, Thailand
| | - Jatuporn Ngoenkam
- Department
of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Pussadee Paensuwan
- Department
of Optometry, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sutatip Pongcharoen
- Department
of Medicine, Faculty of Medicine, Naresuan
University, Phitsanulok 65000, Thailand
| | - Thishana Singh
- School
of Chemistry and Physics, University of
Kwazulu-Natal, Private Bag
X54001, Durban 4000, South Africa
| | - Puttinan Meepowpan
- Department
of Chemistry, Faculty of Science, Chiang
Mai University, Chiang
Mai 50200, Thailand
- Center
of Excellence in Material Science and Technology, Chiang Mai University, Chiang
Mai 50200, Thailand
| | - Jira Jongcharoenkamol
- Department
of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical
Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
9
|
Xia XH, Liang N, Ma XY, Qin L, Wang SY, Chang ZJ. Inhibition of the NF-κB signaling pathway affects gonadal differentiation and leads to male bias in Paramisgurnus dabryanus. Theriogenology 2023; 207:82-95. [PMID: 37269599 DOI: 10.1016/j.theriogenology.2023.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
In recent years, sex-controlled breeding has emerged as an effective strategy to enhance the yields of economic animals with different growth characteristics, while increasing the economic benefits of aquaculture. It is known that the NF-κB pathway participates in gonadal differentiation and reproduction. Therefore, we used the large-scale loach as a research model for the present study and selected an effective inhibitor of the NF-κB signaling pathway (QNZ). This, to investigates the impacts of the NF-κB signaling pathway on gonadal differentiation during a critical period of gonad development and after maturation. Simultaneously, the sex ratio bias and the reproductive capacities of adult fish were analyzed. Our results indicated that the inhibition of the NF-κB signaling pathway influenced the expression of genes related to gonad development, regulated the gene expression related to the brain-gonad-liver axis of juvenile loaches, and finally impacted the gonadal differentiation of the large-scale loach and promoted a male-biased sex ratio. Meanwhile, high QNZ concentrations affected the reproductive abilities of adult loaches and inhibited the growth performance of offspring. Thus, our results deepened the exploration of sex control in fish and provided a certain research basis for the sustainable development of the aquaculture industry.
Collapse
Affiliation(s)
- Xiao-Hua Xia
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Ning Liang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Xiao-Yu Ma
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Lu Qin
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Song-Yun Wang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Zhong-Jie Chang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| |
Collapse
|
10
|
Hiestand L, Shen S, Sloan W, Nasiri H, Lashley D, Kerscher O. Chemical Genetics Screen of EVP4593 Sensitivity in Budding Yeast Identifies Effects on Mitochondrial Structure and Function. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000806. [PMID: 37179969 PMCID: PMC10170316 DOI: 10.17912/micropub.biology.000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Mitochondria are essential eukaryotic organelles. Mitochondrial dysfunction can lead to mitochondrial myopathies and may contribute to neurodegenerative diseases, cancer, and diabetes. EVP4593, a 6-aminoquinazoline derivative with therapeutic potential, has been shown to inhibit NADH-ubiquinone oxidoreductase (Complex I) of the mitochondrial electron transport chain, causing the release of reactive oxygen species (ROS) and a reduction in ATP synthesis. In isolated mitochondria, EVP4593 inhibits respiration in the nanomolar range (IC 50 = 14-25 nM). However, other EVP4593-specific effects on biological processes have also been described. Consistent with an effect on mitochondrial function in budding yeast, we find that EVP4593 [>25µM] induces a pronounced growth defect when wildtype cells are grown on a non-fermentable carbon source. This sensitivity to EVP4593 is exacerbated by deletion of PDR5 , an ABC transporter that confers multidrug resistance. To better understand the cellular pathways and processes affected by EVP4593, we conducted a genome-wide chemical genetics screen of the yeast knockout collection. The objective was to identify yeast gene deletion strains that exhibit growth defects when subjected to a sublethal concentration of EVP4593 [15µM]. Our screen identified 21 yeast genes that are required for resistance to 15µM EVP4593 in glycerol-containing media. The genes identified in our screen are functionally involved in several distinct categories including mitochondrial structure and function, translational regulation and nutritional sensing, cellular stress response and detoxification. Additionally, we identified cellular phenotypes associated with the exposure to EVP4593, including changes in mitochondrial structure. In conclusion, our study represents the first genome-wide screen in yeast to identify the genetic pathways and cell-protective mechanisms involved in EVP4593 resistance and reveals that this small molecule inhibitor affects both mitochondrial structure and function.
Collapse
Affiliation(s)
- Lexie Hiestand
- Biology, William & Mary, Williamsburg, Virginia, United States
| | - Stella Shen
- Biology, William & Mary, Williamsburg, Virginia, United States
| | - Willough Sloan
- Chemistry, William & Mary, Williamsburg, Virginia, United States
| | - Hamid Nasiri
- Department of Cellular Microbiology, University Hohenheim, Stuttgart, Germany
| | - Dana Lashley
- Chemistry, William & Mary, Williamsburg, Virginia, United States
| | - Oliver Kerscher
- Biology, William & Mary, Williamsburg, Virginia, United States
| |
Collapse
|
11
|
Jia K, Xiong H, Yuan W, Huang L, Xu J, Lu C, Hu Y, Huang K, Luo Q, Ma J, Lu H. Diflovidazin damages the hematopoietic stem cells to zebrafish embryos via the TLR4/ NF-κB/ p53 pathway. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108672. [PMID: 36893927 DOI: 10.1016/j.fsi.2023.108672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/13/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Exposure to environmental contaminants frequently induces the occurrence of blood diseases, but the underlying molecular mechanisms are scarcely known. The toxicity of Diflovidazin (DFD), a widely used mite-remover, to the blood system of non-target organisms requires urgent elucidation. To investigate the deleterious effects of DFD (2, 2.5, and 3 mg/L) on the development and survive of hematopoietic stem cells (HSCs), the zebrafish model was used in this study. DFD exposure reduced the number of HSCs and their subtypes, including macrophages, neutrophils, thymus T-cells, erythrocytes, and platelets. The significant changes in the abnormal apoptosis and differentiation of HSCs were the major reasons for the reduction in blood cells. Using small-molecule antagonists and p53 morpholino revealed that the NF-κB/p53 pathway was responsible for the apoptosis of HSCs upon DFD exposure. The restoration results attributed to the TLR4 inhibitor and molecular docking showed that the TLR4 protein, which was upstream of NF-κB signaling, played a vital role in DFD toxicology. This study elucidates the role and molecular mechanism of DFD in damaging zebrafish HSCs. It provides a theoretical basis for the occurrence of various blood diseases in zebrafish and other organisms.
Collapse
Affiliation(s)
- Kun Jia
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Haibin Xiong
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Wei Yuan
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Lirong Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jiaxin Xu
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Chen Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ying Hu
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Kaijie Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Qiang Luo
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Huiqiang Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Affiliated Hospital of Jinggangshan University, Ji'an, 343009, China.
| |
Collapse
|
12
|
Wang X, Guo Z, Lin J, Sun K, Wang G, Hou L, Xu J, Guo J, Zhang X, Guo F, Wei Y. Indirubin protects chondrocytes and alleviates OA by inhibiting the MAPK and NF-κB pathways. Int Immunopharmacol 2023; 115:109624. [PMID: 36577158 DOI: 10.1016/j.intimp.2022.109624] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/28/2022]
Abstract
PHARMACOLOGICAL RELEVANCE Indirubin (IR) is a key active ingredient in the traditional Chinese medication QingDai, also called indigo naturalis, which are extensively used in China to treat chronic diseases, such as inflammation and cancer. However, the function of IR in reducing chondrocyte inflammation in osteoarthritis (OA) is still unclear. AIM OF THE STUDY The aim of this research was to examine how IR inhibits arthritis and to highlight some of its cellular-level processes. MATERIALS AND METHODS Chondrocytes from the knee joint of C57 mice were gathered and grown for in vitro tests and used to determine the toxicity of IR toward chondrocytes using a CCK8 kit. Chondrocytes were treated with IL-1β and IR or with IL-1β alone, and western blotting was used to determine the expression levels of inflammatory mediators. Meanwhile, through the identification and examination of pertinent markers via quantitative PCR. By using PCR assays, western blotting, toluidine blue staining and safranin O staining, the expression of proteoglycan (AGG) and type II collagen (collagen II) was investigated. Furthermore, western blotting was used to detect activation of the NF-κB and MAPK signaling pathways, and immunofluorescence was used to detect p65 nuclear translocation. In an in vivo experiment, C57BL/6 mice were subjected to destabilization of the medial meniscus (DMM) surgery to produce an OA model, and IR was injected into the articular cavity for 8 weeks. Eventually, the mice were killed, and samples of the knee joints were obtained for histological examination and analysis. RESULTS IR significantly reduced the expression of inflammatory regulators in chondrocytes treated with IL-1β, including iNOS and COX-2. Inhibition of IL-1β induced production of the key catabolic enzymes MMP3, MMP13 and A5. Additionally, an improvement in the downregulation of collagen II and AGG expression was observed. Moreover, IR prevented the aberrant IL-1β-induced activation of the NF-κB and MAPK signaling pathways, which resulted in downregulation of p65 and p38 expression. Compared to the DMM group, the severity of cartilage injury in animals was dramatically lessened and OARSI scores were lower in the treated groups. CONCLUSION According to the above findings, IR is quite effective in preventing arthritis both in vivo and in vitro, suggesting that IR may be employed as a possible anti-arthritis drug.
Collapse
Affiliation(s)
- Xiaolin Wang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, PR China; Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Zhou Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jiamin Lin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Genchun Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jingting Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jiachao Guo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Xiong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan 430030, Hubei, PR China.
| | - Youxiu Wei
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, PR China.
| |
Collapse
|
13
|
Mailem RC, Tayo LL. Drug Repurposing Using Gene Co-Expression and Module Preservation Analysis in Acute Respiratory Distress Syndrome (ARDS), Systemic Inflammatory Response Syndrome (SIRS), Sepsis, and COVID-19. BIOLOGY 2022; 11:biology11121827. [PMID: 36552336 PMCID: PMC9775208 DOI: 10.3390/biology11121827] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
SARS-CoV-2 infections are highly correlated with the overexpression of pro-inflammatory cytokines in what is known as a cytokine storm, leading to high fatality rates. Such infections are accompanied by SIRS, ARDS, and sepsis, suggesting a potential link between the three phenotypes. Currently, little is known about the transcriptional similarity between these conditions. Herein, weighted gene co-expression network analysis (WGCNA) clustering was applied to RNA-seq datasets (GSE147902, GSE66890, GSE74224, GSE177477) to identify modules of highly co-expressed and correlated genes, cross referenced with dataset GSE160163, across the samples. To assess the transcriptome similarities between the conditions, module preservation analysis was performed and functional enrichment was analyzed in DAVID webserver. The hub genes of significantly preserved modules were identified, classified into upregulated or downregulated, and used to screen candidate drugs using Connectivity Map (CMap) to identify repurposed drugs. Results show that several immune pathways (chemokine signaling, NOD-like signaling, and Th1 and Th2 cell differentiation) are conserved across the four diseases. Hub genes screened using intramodular connectivity show significant relevance with the pathogenesis of cytokine storms. Transcriptomic-driven drug repurposing identified seven candidate drugs (SB-202190, eicosatetraenoic-acid, loratadine, TPCA-1, pinocembrin, mepacrine, and CAY-10470) that targeted several immune-related processes. These identified drugs warrant further study into their efficacy for treating cytokine storms, and in vitro and in vivo experiments are recommended to confirm the findings of this study.
Collapse
Affiliation(s)
- Ryan Christian Mailem
- School of Chemical, Biological, and Materials Engineering and Sciences and School of Graduate Studies, Mapúa University, Manila City 1002, Philippines
| | - Lemmuel L. Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences and School of Graduate Studies, Mapúa University, Manila City 1002, Philippines
- School of Health Sciences, Mapúa University, Manila City 1002, Philippines
- Correspondence: ; Tel.: +63-02-247-5000 (ext. 3300)
| |
Collapse
|
14
|
Grekhnev DA, Kruchinina AA, Vigont VA, Kaznacheyeva EV. The Mystery of EVP4593: Perspectives of the Quinazoline-Derived Compound in the Treatment of Huntington's Disease and Other Human Pathologies. Int J Mol Sci 2022; 23:15724. [PMID: 36555369 PMCID: PMC9778905 DOI: 10.3390/ijms232415724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Quinazoline derivatives have various pharmacological activities and are widely used in clinical practice. Here, we reviewed the proposed mechanisms of the physiological activity of the quinazoline derivative EVP4593 and perspectives for its clinical implication. We summarized the accumulated data about EVP4593 and focused on its activities in different models of Huntington's disease (HD), including patient-specific iPSCs-based neurons. To make a deeper insight into its neuroprotective role in HD treatment, we discussed the ability of EVP4593 to modulate calcium signaling and reduce the level of the huntingtin protein. Moreover, we described possible protective effects of EVP4593 in other pathologies, such as oncology, cardiovascular diseases and parasite invasion. We hope that comprehensive analyses of the molecular mechanisms of EVP4593 activity will allow for the expansion of the scope of the EVP4593 application.
Collapse
|
15
|
Kurelac I, Cavina B, Sollazzo M, Miglietta S, Fornasa A, De Luise M, Iorio M, Lama E, Traversa D, Nasiri HR, Ghelli A, Musiani F, Porcelli AM, Iommarini L, Gasparre G. NDUFS3 knockout cancer cells and molecular docking reveal specificity and mode of action of anti-cancer respiratory complex I inhibitors. Open Biol 2022; 12:220198. [PMID: 36349549 PMCID: PMC9653258 DOI: 10.1098/rsob.220198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Inhibition of respiratory complex I (CI) is becoming a promising anti-cancer strategy, encouraging the design and the use of inhibitors, whose mechanism of action, efficacy and specificity remain elusive. As CI is a central player of cellular bioenergetics, a finely tuned dosing of targeting drugs is required to avoid side effects. We compared the specificity and mode of action of CI inhibitors metformin, BAY 87-2243 and EVP 4593 using cancer cell models devoid of CI. Here we show that both BAY 87-2243 and EVP 4593 were selective, while the antiproliferative effects of metformin were considerably independent from CI inhibition. Molecular docking predictions indicated that the high efficiency of BAY 87-2243 and EVP 4593 may derive from the tight network of bonds in the quinone binding pocket, although in different sites. Most of the amino acids involved in such interactions are conserved across species and only rarely found mutated in human. Our data make a case for caution when referring to metformin as a CI-targeting compound, and highlight the need for dosage optimization and careful evaluation of molecular interactions between inhibitors and the holoenzyme.
Collapse
Affiliation(s)
- Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Beatrice Cavina
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Stefano Miglietta
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Agnese Fornasa
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Maria Iorio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Eleonora Lama
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Daniele Traversa
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Hamid Razi Nasiri
- Department of Cellular Microbiology, University Hohenheim, Stuttgart, Germany
| | - Anna Ghelli
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy,Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Francesco Musiani
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy,Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy,Interdepartmental Centre for Industrial Research ‘Scienze della Vita e Tecnologie per la Salute’, University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy,Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy,Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Dixon EE, Bradford STJ, Humphreys BD. Mini kidney organoids deliver maximal drug screening impact. Cell Stem Cell 2022; 29:1011-1012. [PMID: 35803221 DOI: 10.1016/j.stem.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In this issue of Cell Stem Cell, Tran and colleagues develop a platform for differentiating thousands of miniature kidney organoids consisting of one or two nephron-like structures each. They use this platform to identify a potent new inhibitor of cyst growth in organoid models of autosomal-dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Eryn E Dixon
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Shayna T J Bradford
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA; Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
17
|
Tran T, Song CJ, Nguyen T, Cheng SY, McMahon JA, Yang R, Guo Q, Der B, Lindström NO, Lin DCH, McMahon AP. A scalable organoid model of human autosomal dominant polycystic kidney disease for disease mechanism and drug discovery. Cell Stem Cell 2022; 29:1083-1101.e7. [PMID: 35803227 PMCID: PMC11088748 DOI: 10.1016/j.stem.2022.06.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/28/2022] [Accepted: 06/08/2022] [Indexed: 12/13/2022]
Abstract
Human pluripotent stem-cell-derived organoids are models for human development and disease. We report a modified human kidney organoid system that generates thousands of similar organoids, each consisting of 1-2 nephron-like structures. Single-cell transcriptomic profiling and immunofluorescence validation highlighted patterned nephron-like structures utilizing similar pathways, with distinct morphogenesis, to human nephrogenesis. To examine this platform for therapeutic screening, the polycystic kidney disease genes PKD1 and PKD2 were inactivated by gene editing. PKD1 and PKD2 mutant models exhibited efficient and reproducible cyst formation. Cystic outgrowths could be propagated for months to centimeter-sized cysts. To shed new light on cystogenesis, 247 protein kinase inhibitors (PKIs) were screened in a live imaging assay identifying compounds blocking cyst formation but not overall organoid growth. Scaling and further development of the organoid platform will enable a broader capability for kidney disease modeling and high-throughput drug screens.
Collapse
Affiliation(s)
- Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Cheng Jack Song
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Amgen Research, Cardiometabolic Disorders, 1120 Veterans Blvd, South San Francisco, CA 94080, USA
| | - Trang Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Shun-Yang Cheng
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Rui Yang
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Balint Der
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel C-H Lin
- Amgen Research, Cardiometabolic Disorders, 1120 Veterans Blvd, South San Francisco, CA 94080, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
18
|
Human Herpesvirus 6A Tegument Protein U14 Induces NF-κB Signaling by Interacting with p65. J Virol 2021; 95:e0126921. [PMID: 34549982 DOI: 10.1128/jvi.01269-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral infection induces host cells to mount a variety of immune responses, which may either limit viral propagation or create conditions conducive to virus replication in some instances. In this regard, activation of the NF-κB transcription factor is known to modulate virus replication. Human herpesvirus 6A (HHV-6A), which belongs to the Betaherpesvirinae subfamily, is frequently found in patients with neuroinflammatory diseases, although its role in disease pathogenesis has not been elucidated. In this study, we found that the HHV-6A-encoded U14 protein activates NF-κB signaling following interaction with the NF-κB complex protein, p65. Through induction of nuclear translocation of p65, U14 increases the expression of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein 1 transcripts. We also demonstrated that activation of NF-κB signaling is important for HHV-6A replication, since inhibition of this pathway reduced virus protein accumulation and viral genome copy number. Taken together, our results suggest that HHV-6A infection activates the NF-κB pathway and promotes viral gene expression via late gene products, including U14. IMPORTANCE Human herpesvirus 6A (HHV-6A) is frequently found in patients with neuro-inflammation, although its role in the pathogenesis of this disease has not been elucidated. Most viral infections activate the NF-κB pathway, which causes the transactivation of various genes, including those encoding proinflammatory cytokines. Our results indicate that HHV-6A U14 activates the NF-κB pathway, leading to upregulation of proinflammatory cytokines. We also found that activation of the NF-κB transcription factor is important for efficient viral replication. This study provides new insight into HHV-6A U14 function in host cell signaling and identifies potential cellular targets involved in HHV-6A pathogenesis and replication.
Collapse
|
19
|
New targets of morphine postconditioning protection of the myocardium in ischemia/reperfusion injury: Involvement of HSP90/Akt and C5a/NF-κB. Open Med (Wars) 2021; 16:1552-1563. [PMID: 34722891 PMCID: PMC8525660 DOI: 10.1515/med-2021-0340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/21/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
Abstract
Background Activation of the complement component 5a (C5a) and nuclear factor κB (NF-κB) signaling is an important feature of myocardial ischemia/reperfusion (I/R) injury and recent studies show that morphine postconditioning (MP) attenuates the myocardial injury. However, the mediating cardioprotective mechanisms remain unclear. The present study explores the role and interaction of heat shock protein 90 (HSP90), Akt, C5a, and NF-κB in MP-induced cardioprotection. Methods Male Sprague Dawley rats (n = 160) were randomized into eight groups (n = 20 per group). Rats in the sham group underwent thoracotomy, passing the ligature through the heart but without tying it (150 min), and the other seven groups were subjected to 30 min of anterior descending coronary artery occlusion followed by 2 h of reperfusion and the following treatments: I/R (30 min of ischemia and followed by 2 h of reperfusion); ischemic postconditioning (IPostC, 30 s of ischemia altered with 30 s of reperfusion, repeated for three cycles, and followed by reperfusion for 2 h); MP (0.3 mg/kg morphine administration 10 min before reperfusion); MP combined with the HSP90 inhibitor geldanamycin (GA, 1 mg/kg); MP combined with the Akt inhibitor GSK-690693 (GSK, 20 mg/kg); and MP combined with the C5a inhibitor PMX205 (PMX, 1 mg/kg/day, administration via drinking water for 28 days) and MP combined with the NF-κB inhibitor EVP4593 (QNZ, 1 mg/kg). All inhibitors were administered 10 min before morphine and followed by 2 h reperfusion. Results MP significantly reduced the I/R-induced infarct size, the apoptosis, and the release of cardiac troponin I, lactate dehydrogenase (LDH), and creatine kinase-MB. These beneficial effects were accompanied by increased expression of HSP90 and p-Akt, and decreased expression of C5a, NF-κB, tumor necrosis factor α, interleukin-1β, and intercellular cell adhesion molecule 1. However, HSP90 inhibitor GA or Akt inhibitor GSK increased the expression of C5a and NF-κB and prevented MP-induced cardioprotection. Furthermore, GA inhibited the MP-induced upregulation of p-Akt, while GSK did not affect HSP90, indicating that p-Akt acts downstream of HSP90 in MP-induced cardioprotection. In addition, C5a inhibitor PMX enhanced the MP-induced downregulation of NF-κB, while NF-κB inhibitor QNZ had no effect on C5a, indicating that the C5a/NF-κB signaling pathway is involved in MP-induced cardioprotection. Conclusion HSP90 is critical for MP-mediated cardioprotection possibly by promoting the phosphorylation of Akt and inhibiting the activation of C5a and NF-κB signaling and the subsequent myocardial inflammation, ultimately attenuating the infarct size and cardiomyocyte apoptosis.
Collapse
|
20
|
Syntheses and Structure-Activity Relationships of N-Phenethyl-Quinazolin-4-yl-Amines as Potent Inhibitors of Cytochrome bd Oxidase in Mycobacterium tuberculosis. APPLIED SCIENCES (BASEL, SWITZERLAND) 2021; 11:9092. [PMID: 36698770 PMCID: PMC9873234 DOI: 10.3390/app11199092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The development of cytochrome bd oxidase (cyt-bd) inhibitors are needed for comprehensive termination of energy production in Mycobacterium tuberculosis (Mtb) to treat tuberculosis infections. Herein, we report on the structure-activity-relationships (SAR) of 22 new N-phenethyl-quinazolin-4-yl-amines that target cyt-bd. Our focused set of compounds was synthesized and screened against three mycobacterial strains: Mycobacterium bovis BCG, Mycobacterium tuberculosis H37Rv and the clinical isolate Mycobacterium tuberculosis N0145 with and without the cytochrome bcc:aa 3 inhibitor Q203 in an ATP depletion assay. Two compounds, 12a and 19a, were more active against all three strains than the naturally derived cyt-bd inhibitor aurachin D.
Collapse
|
21
|
Wang L, Zhang Q, Wang Z, Zhu W, Tan N. Design, synthesis, docking, molecular dynamics and bioevaluation studies on novel N-methylpicolinamide and thienopyrimidine derivatives with inhibiting NF-κB and TAK1 activities: Cheminformatics tools RDKit applied in drug design. Eur J Med Chem 2021; 223:113576. [PMID: 34153577 DOI: 10.1016/j.ejmech.2021.113576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/26/2022]
Abstract
Using cheminformatics tools RDKit and literature investigation, four series of 24 thienopyrimidine/N-methylpicolinamide derivatives substituted with pyrimidine were designed, synthesized and evaluated for activities against three cancer cell lines (MDA-MB-231, HCT116 and A549), TAK1 kinase and NF-κB signaling pathway. Almost all compounds showed selectivity toward the A549 cell lines and the most promising compound 38 could inhibit TAK1 kinase and NF-κB signaling pathway with the IC50 values of 0.58 and 0.84 μM. Moreover, 38 can induce cell cycle arrest of A549 cells at the G2/M checkpoint with 30.57% and induce apoptosis (34.94%) in a concentration-dependent manner. And western blot showed that compound 38 could inhibit TNF-α-induced IκBα phosphorylation, IκBα degradation, p65 phosphorylation and TAK1 phosphorylation, and reduce the expression of p65. What's more, the studies of docking, molecular dynamics, MM/PBSA and frequency analysis theoretically supported the conclusions of the bioevaluation.
Collapse
Affiliation(s)
- Linxiao Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Qian Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science &Technology Normal University, Nanchang, 330013, China.
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
22
|
Synthesis, crystal structure, spectroscopic and photophysical studies of novel fluorinated quinazoline derivatives. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.129951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Malarz K, Mularski J, Kuczak M, Mrozek-Wilczkiewicz A, Musiol R. Novel Benzenesulfonate Scaffolds with a High Anticancer Activity and G2/M Cell Cycle Arrest. Cancers (Basel) 2021; 13:cancers13081790. [PMID: 33918637 PMCID: PMC8068801 DOI: 10.3390/cancers13081790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Sulfonates, unlike their derivatives, sulphonamides, have rarely been investigated for their anticancer activity. Unlike the well-known sulphonamides, esters are mainly used as convenient intermediates in a synthesis. Here, we present the first in-depth investigation of quinazoline sulfonates. A small series of derivatives were synthesized and tested for their anticancer activity. Based on their structural similarity, these compounds resemble tyrosine kinase inhibitors and the p53 reactivator CP-31398. Their biological activity profile, however, was more related to sulphonamides because there was a strong cell cycle arrest in the G2/M phase. Further investigation revealed a multitargeted mechanism of the action that corresponded to the p53 protein status in the cell. Although the compounds expressed a high submicromolar activity against leukemia and colon cancers, pancreatic cancer and glioblastoma were also susceptible. Apoptosis and autophagy were confirmed as the cell death modes that corresponded with the inhibition of metabolic activity and the activation of the p53-dependent and p53-independent pathways. Namely, there was a strong activation of the p62 protein and GADD44. Other proteins such as cdc2 were also expressed at a higher level. Moreover, the classical caspase-dependent pathway in leukemia was observed at a lower concentration, which again confirmed a multitargeted mechanism. It can therefore be concluded that the sulfonates of quinazolines can be regarded as promising scaffolds for developing anticancer agents.
Collapse
Affiliation(s)
- Katarzyna Malarz
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
- Correspondence: (K.M.); (R.M.)
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
| | - Michał Kuczak
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
| | - Anna Mrozek-Wilczkiewicz
- A. Chełkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland; (M.K.); (A.M.-W.)
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland;
- Correspondence: (K.M.); (R.M.)
| |
Collapse
|
24
|
Wei X, Li C, Zhang Y, Li K, Li J, Ai K, Li K, Zhang J, Yang J. Fish NF‐κB couples TCR and IL‐17 signals to regulate ancestral T‐cell immune response against bacterial infection. FASEB J 2021; 35:e21457. [DOI: 10.1096/fj.202002393rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Cheng Li
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Yu Zhang
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Jiaqi Li
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Kete Ai
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Kunming Li
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research School of Life Sciences East China Normal University Shanghai China
- Laboratory for Marine Biology and Biotechnology Qingdao National Laboratory for Marine Science and Technology Qingdao China
| |
Collapse
|
25
|
A transposon screen identifies enhancement of NF-κB pathway as a mechanism of resistance to eribulin. Breast Cancer 2021; 28:884-895. [PMID: 33616862 DOI: 10.1007/s12282-021-01224-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/07/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Eribulin mesylate (eribulin) is an efficient microtubule inhibitor that is used for metastatic breast cancer. However, breast cancer can develop resistance to eribulin. This resistance mechanism needs to be elucidated. METHODS A transposon mutagenesis screen was conducted using a pPB-SB-CMV-puro-SD plasmid and pCMV-PBase transposase. Viability and cytotoxicity were analyzed by MTT assay and flow cytometry, respectively. Real-time PCR and western blot were used for gene expression analysis. In addition, vivo study was also designed to analyze therapy efficiency. RESULTS TAB2, which is part of the nuclear factor-kappa B (NF-κB) pathway, was identified as a candidate eribulin-resistant gene. TAB2 down-regulation resulted in significantly lower cell viability and higher cytotoxicity of cells treated with eribulin, while TAB2 up-regulation showed opposite results. Similarly, combination of NF-κB inhibitors [Bay-117082 and QNZ (quinazoline derivative)] with eribulin showed significantly lower cell viability and higher drug cytotoxicity than single agent treatment with eribulin in MDA-MB-231 cells. However, QNZ increased NF-κB activity in MCF7 cells by up-regulating TAB2, which reduced the sensitivity to eribulin. Furthermore, combination of Bay-117082 with eribulin induced greater regression of MDA-MB-231 tumors compared to eribulin monotherapy in vivo. CONCLUSIONS These results consistently illustrated that TAB2-NF-κB pathway may increases resistance to eribulin in breast cancer models. Moreover, these results support the use of a combination strategy of eribulin with NF-κB inhibitors, and provide evidence that transposon mutagenesis screens are capable of identifying drug-resistant genes.
Collapse
|
26
|
Elsocht M, Giron P, Maes L, Versées W, Gutierrez GJ, De Grève J, Ballet S. Structure-Activity Relationship (SAR) Study of Spautin-1 to Entail the Discovery of Novel NEK4 Inhibitors. Int J Mol Sci 2021; 22:ijms22020635. [PMID: 33435251 PMCID: PMC7827406 DOI: 10.3390/ijms22020635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 11/16/2022] Open
Abstract
Lung cancer is one of the most frequently diagnosed cancers accounting for the highest number of cancer-related deaths in the world. Despite significant progress including targeted therapies and immunotherapy, the treatment of advanced lung cancer remains challenging. Targeted therapies are highly efficacious at prolonging life, but not curative. In prior work we have identified Ubiquitin Specific Protease 13 (USP13) as a potential target to significantly enhance the efficacy of mutant EGFR inhibition. The current study aimed to develop lead molecules for the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) by developing potent USP13 inhibitors initially starting from Spautin-1, the only available USP13 inhibitor. A SAR study was performed which revealed that increasing the chain length between the secondary amine and phenyl group and introducing a halogen capable of inducing a halogen bond at position 4' of the phenyl group, dramatically increased the activity. However, we could not confirm the binding between Spautin-1 (or its analogues) and USP13 using isothermal titration calorimetry (ITC) or thermal shift assay (TSA) but do not exclude binding under physiological conditions. Nevertheless, we found that the anti-proliferative activity displayed by Spautin-1 towards EGFR-mutant NSCLC cells in vitro was at least partially associated with kinase inhibition. In this work, we present N-[2-(substituted-phenyl)ethyl]-6-fluoro-4-quinazolinamines as promising lead compounds for the treatment of NSCLC. These analogues are significantly more effective towards EGFR-mutant NSCLC cells than Spautin-1 and act as potent never in mitosis A related kinase 4 (NEK4) inhibitors (IC50~1 µM) with moderate selectivity over other kinases.
Collapse
Affiliation(s)
- Mathias Elsocht
- Research Group of Organic Chemistry, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium;
| | - Philippe Giron
- Laboratory of Medical and Molecular Oncology and Center of Medical Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (P.G.); (J.D.G.)
- Laboratory of Pathophysiological Cell Signalling (PACS), Department of Biology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium;
| | - Laila Maes
- VIB-VUB Center for Structural Biology, Pleinlaan 2, 1050 Brussels, Belgium; (L.M.); (W.V.)
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Wim Versées
- VIB-VUB Center for Structural Biology, Pleinlaan 2, 1050 Brussels, Belgium; (L.M.); (W.V.)
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Gustavo J. Gutierrez
- Laboratory of Pathophysiological Cell Signalling (PACS), Department of Biology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium;
| | - Jacques De Grève
- Laboratory of Medical and Molecular Oncology and Center of Medical Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (P.G.); (J.D.G.)
| | - Steven Ballet
- Research Group of Organic Chemistry, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium;
- Correspondence: ; Tel.: +32-2-6293292
| |
Collapse
|
27
|
Prikhodko V, Chernyuk D, Sysoev Y, Zernov N, Okovityi S, Popugaeva E. Potential Drug Candidates to Treat TRPC6 Channel Deficiencies in the Pathophysiology of Alzheimer's Disease and Brain Ischemia. Cells 2020; 9:cells9112351. [PMID: 33114455 PMCID: PMC7692306 DOI: 10.3390/cells9112351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer’s disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer’s disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer’s disease and cerebral ischemia.
Collapse
Affiliation(s)
- Veronika Prikhodko
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Daria Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Yurii Sysoev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Sergey Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Correspondence:
| |
Collapse
|
28
|
Hunt DWC, Ivanova IA, Dagnino L. DRM02, a novel phosphodiesterase-4 inhibitor with cutaneous anti-inflammatory activity. Tissue Barriers 2020; 8:1765633. [PMID: 32479135 DOI: 10.1080/21688370.2020.1765633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic inflammatory skin disorders are frequently associated with impaired skin barrier function. Selective phosphodiesterase-4 (PDE4) inhibition constitutes an effective therapeutic strategy for the treatment of inflammatory skin diseases. We now report the pharmacological anti-inflammatory profile of DRM02, a novel pyrazolylbenzothiazole derivative with selective in vitro inhibitory activity toward PDE4 isoforms A, B and D. DRM02 treatment of cultured primary human and mouse epidermal keratinocytes interfered with pro-inflammatory cytokine production elicited by interleukin-1α and tumor necrosis factor-α. Similarly, DRM02 inhibited the production of pro-inflammatory cytokines by human peripheral blood mononuclear cells ex vivo and cultured THP-1 monocyte-like cells, with IC50 values of 0.6-14 µM. These anti-inflammatory properties of DRM02 were associated with dose-dependent repression of nuclear factor-κB (NF-κB) transcriptional activity. In skin inflammation in vivo mouse models, topically applied DRM02 inhibited the acute response to phorbol ester and induced Th2-type contact hypersensitivity reactivity. Further, DRM02 also decreased cutaneous clinical changes and expression of Th17 immune pathway cytokines in a mouse model of psoriasis evoked by repeated topical imiquimod application. Thus, the overall pharmacological profiling of the PDE4 inhibitor DRM02 has revealed its potential as a topical therapy for inflammatory skin disorders and restoration of skin homeostasis.
Collapse
Affiliation(s)
| | - Iordanka A Ivanova
- Department of Physiology and Pharmacology, University of Western Ontario , London, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, University of Western Ontario , London, Canada.,Department of Oncology, University of Western Ontario , London, Canada
| |
Collapse
|
29
|
Abstract
Afatinib is a 4-anilinoquinazoline tyrosine kinase inhibitor (TKI) in the form of a dimaleate salt which is indicated for the treatment of locally advanced or metastatic non-small cell lung cancer (NSCLC). The most scalable route for the synthesis of this drug was reported in two Boehringer Ingelheim patents, in which the title compound, 4,7-dichloro-6-nitroquinazoline (IV), is an important intermediate. Compound IV is also present in a number of synthetic pathways for various 4,7-disubstituted quinazoline derivatives displaying high therapeutic potential. However, no detailed characterization of this popular compound has been reported, possibly due to its high instability. In this paper, IV was prepared in an overall yield of 56.1% by a 3-step process (condensation, nitration, and chlorination) from 2-amino-4-chlorobenzoic acid (I). The target compound has been for the first time fully characterized by melting point, mass-spectrometry, FT-IR, 1H-NMR and 13C-NMR spectroscopies.
Collapse
|
30
|
Yang Y, Delalio LJ, Best AK, Macal E, Milstein J, Donnelly I, Miller AM, McBride M, Shu X, Koval M, Isakson BE, Johnstone SR. Endothelial Pannexin 1 Channels Control Inflammation by Regulating Intracellular Calcium. THE JOURNAL OF IMMUNOLOGY 2020; 204:2995-3007. [PMID: 32312847 PMCID: PMC7336877 DOI: 10.4049/jimmunol.1901089] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/27/2020] [Indexed: 12/26/2022]
Abstract
The proinflammatory cytokine IL-1β is a significant risk factor in cardiovascular disease that can be targeted to reduce major cardiovascular events. IL-1β expression and release are tightly controlled by changes in intracellular Ca2+ ([Ca2+]i), which has been associated with ATP release and purinergic signaling. Despite this, the mechanisms that regulate these changes have not been identified. The pannexin 1 (Panx1) channels have canonically been implicated in ATP release, especially during inflammation. We examined Panx1 in human umbilical vein endothelial cells following treatment with the proinflammatory cytokine TNF-α. Analysis by whole transcriptome sequencing and immunoblot identified a dramatic increase in Panx1 mRNA and protein expression that is regulated in an NF-κB-dependent manner. Furthermore, genetic inhibition of Panx1 reduced the expression and release of IL-1β. We initially hypothesized that increased Panx1-mediated ATP release acted in a paracrine fashion to control cytokine expression. However, our data demonstrate that IL-1β expression was not altered after direct ATP stimulation in human umbilical vein endothelial cells. Because Panx1 forms a large pore channel, we hypothesized it may permit Ca2+ diffusion into the cell to regulate IL-1β. High-throughput flow cytometric analysis demonstrated that TNF-α treatments lead to elevated [Ca2+]i, corresponding with Panx1 membrane localization. Genetic or pharmacological inhibition of Panx1 reduced TNF-α-associated increases in [Ca2+]i, blocked phosphorylation of the NF-κB-p65 protein, and reduced IL-1β transcription. Taken together, the data in our study provide the first evidence, to our knowledge, that [Ca2+]i regulation via the Panx1 channel induces a feed-forward effect on NF-κB to regulate IL-1β synthesis and release in endothelium during inflammation.
Collapse
Affiliation(s)
- Yang Yang
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908.,Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Leon J Delalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Angela K Best
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Edgar Macal
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Jenna Milstein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Iona Donnelly
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Ashley M Miller
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Martin McBride
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Xiaohong Shu
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322.,Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322; and
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908; .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Scott R Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908;
| |
Collapse
|
31
|
Funsten JR, Murillo Brizuela KO, Swatzel HE, Ward AS, Scott TA, Eikenbusch SM, Shields MC, Meredith JL, Mitchell TY, Hanna ML, Bingham KN, Rawlings JS. PKC signaling contributes to chromatin decondensation and is required for competence to respond to IL-2 during T cell activation. Cell Immunol 2020; 347:104027. [PMID: 31864664 PMCID: PMC10731676 DOI: 10.1016/j.cellimm.2019.104027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022]
Abstract
The clonal proliferation of antigen-specific T cells during an immune response critically depends on the differential response to growth factors, such as IL-2. While activated T cells proliferate robustly in response to IL-2 stimulation, naïve (quiescent) T cells are able to ignore the potent effects of growth factors because they possess chromatin that is tightly condensed such that transcription factors, such as STAT5, cannot access DNA. Activation via the T cell receptor (TCR) induces a rapid decondensation of chromatin, permitting STAT5-DNA engagement and ultimately promoting proliferation of only antigen-specific T cells. Previous work demonstrated that the mobilization of intracellular calcium following TCR stimulation is a key event in the decondensation of chromatin. Here we examine PKC-dependent signaling mechanisms to determine their role in activation-induced chromatin decondensation and the subsequent acquisition of competence to respond to IL-2 stimulation. We found that a calcium-dependent PKC contributes to activation-induced chromatin decondensation and that the p38 MAPK and NFκB pathways downstream of PKC each contribute to regulating the proper decondensation of chromatin. Importantly, we found that p44/42 MAPK activity is required for peripheral T cells to gain competence to properly respond to IL-2 stimulation. Our findings shed light on the mechanisms that control the clonal proliferation of antigen-specific peripheral T cells during an immune response.
Collapse
Affiliation(s)
| | | | - Hayley E Swatzel
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Audrey S Ward
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Tia A Scott
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | | | - Molly C Shields
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Jenna L Meredith
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | | | - Megan L Hanna
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Kellie N Bingham
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Jason S Rawlings
- Department of Biology, Furman University, Greenville, SC 29613, USA.
| |
Collapse
|
32
|
Khusbu FY, Zhou X, Roy M, Chen FZ, Cao Q, Chen HC. Resveratrol induces depletion of TRAF6 and suppresses prostate cancer cell proliferation and migration. Int J Biochem Cell Biol 2019; 118:105644. [PMID: 31712163 DOI: 10.1016/j.biocel.2019.105644] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/08/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
Although the early diagnosis of prostate cancer (PCa) enhances life expectancy with a 5-year survival rate of 100 %, metastasized-PCa is the fundamental reason for death by PCa, hence requires an advanced and target-directed treatment strategy. Metastasis is considered to be initiated with the epithelial-mesenchymal transition (EMT) event in which tumor cells change their epithelial characteristics into mesenchymal form and exacerbates the cancer progression. Herein, we investigated the effect and mechanism of resveratrol function in PCa cell proliferation and migration and reported that TNF-receptor associated factor 6 (TRAF6), an unconventional E3 ligase, is a key mediator of resveratrol function to inhibit PCa cell growth and proliferation and targeted for lysosomal degradation by resveratrol. MTT and cell counting demonstrated that resveratrol inhibited the viability and proliferation in DU145 and PC3 cells. Resveratrol (50 μM) mediated the degradation of TRAF6 which in turn facilitated repression of the NF-κB pathway. Also, wound healing and transwell migration assays and level of EMT-related proteins showed that resveratrol used TRAF6, at least in part to inhibit cell migration. Overexpression of TRAF6 augmented EMT in PCa by upregulating the expression of transcription factor SLUG. Moreover, TRAF6 overexpression was closely associated with EMT process through the NF-κB pathway. Our exploration exhibited that resveratrol may inhibit EMT through the TRAF6/NF-κB/SLUG axis. Altogether, this study represents that TRAF6 acts as an intermediary of resveratrol action to suppress PCa cell proliferation and migration, and concerns future attention to obtain as a therapeutic target for the treatment of PCa.
Collapse
Affiliation(s)
- Farjana Yeasmin Khusbu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China.
| | - Xi Zhou
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Mridul Roy
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Fang-Zhi Chen
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Qian Cao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Han-Chun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
33
|
The Innate Immune Sensor NLRC3 Acts as a Rheostat that Fine-Tunes T Cell Responses in Infection and Autoimmunity. Immunity 2019; 49:1049-1061.e6. [PMID: 30566882 DOI: 10.1016/j.immuni.2018.10.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/12/2018] [Accepted: 10/03/2018] [Indexed: 12/31/2022]
Abstract
Appropriate immune responses require a fine balance between immune activation and attenuation. NLRC3, a non-inflammasome-forming member of the NLR innate immune receptor family, attenuates inflammation in myeloid cells and proliferation in epithelial cells. T lymphocytes express the highest amounts of Nlrc3 transcript where its physiologic relevance is unknown. We show that NLRC3 attenuated interferon-γ and TNF expression by CD4+ T cells and reduced T helper 1 (Th1) and Th17 cell proliferation. Nlrc3-/- mice exhibited increased and prolonged CD4+ T cell responses to lymphocytic choriomeningitis virus infection and worsened experimental autoimmune encephalomyelitis (EAE). These functions of NLRC3 were executed in a T-cell-intrinsic fashion: NLRC3 reduced K63-linked ubiquitination of TNF-receptor-associated factor 6 (TRAF6) to limit NF-κB activation, lowered phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), and diminished glycolysis and oxidative phosphorylation. This study reveals an unappreciated role for NLRC3 in attenuating CD4+ T cell signaling and metabolism.
Collapse
|
34
|
NOD-like receptors: major players (and targets) in the interface between innate immunity and cancer. Biosci Rep 2019; 39:BSR20181709. [PMID: 30837326 PMCID: PMC6454022 DOI: 10.1042/bsr20181709] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022] Open
Abstract
Innate immunity comprises several inflammation-related modulatory pathways which receive signals from an array of membrane-bound and cytoplasmic pattern recognition receptors (PRRs). The NLRs (NACHT (NAIP (neuronal apoptosis inhibitor protein), C2TA (MHC class 2 transcription activator), HET-E (incompatibility locus protein from Podospora anserina) and TP1 (telomerase-associated protein) and Leucine-Rich Repeat (LRR) domain containing proteins) relate to a large family of cytosolic innate receptors, involved in detection of intracellular pathogens and endogenous byproducts of tissue injury. These receptors may recognize pathogen-associated molecular patterns (PAMPs) and/or danger-associated molecular patterns (DAMPs), activating host responses against pathogen infection and cellular stress. NLR-driven downstream signals trigger a number of signaling circuitries, which may either initiate the formation of inflammasomes and/or activate nuclear factor κB (NF-κB), stress kinases, interferon response factors (IRFs), inflammatory caspases and autophagy. Disruption of those signals may lead to a number of pro-inflammatory conditions, eventually promoting the onset of human malignancies. In this review, we describe the structures and functions of the most well-defined NLR proteins and highlight their association and biological impact on a diverse number of cancers.
Collapse
|
35
|
Shariati M, Imanzadeh G, Rostami A, Ghoreishy N, Kheirjou S. Application of laccase/DDQ as a new bioinspired catalyst system for the aerobic oxidation of tetrahydroquinazolines and Hantzsch 1,4-dihydropyridines. CR CHIM 2019. [DOI: 10.1016/j.crci.2019.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
36
|
Scheurer MJJ, Brands RC, El-Mesery M, Hartmann S, Müller-Richter UDA, Kübler AC, Seher A. The Selection of NFκB Inhibitors to Block Inflammation and Induce Sensitisation to FasL-Induced Apoptosis in HNSCC Cell Lines Is Critical for Their Use as a Prospective Cancer Therapy. Int J Mol Sci 2019; 20:ijms20061306. [PMID: 30875877 PMCID: PMC6471923 DOI: 10.3390/ijms20061306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 02/02/2023] Open
Abstract
Inflammation is a central aspect of tumour biology and can contribute significantly to both the origination and progression of tumours. The NFκB pathway is one of the most important signal transduction pathways in inflammation and is, therefore, an excellent target for cancer therapy. In this work, we examined the influence of four NFκB inhibitors—Cortisol, MLN4924, QNZ and TPCA1—on proliferation, inflammation and sensitisation to apoptosis mediated by the death ligand FasL in the HNSCC cell lines PCI1, PCI9, PCI13, PCI52 and SCC25 and in the human dermal keratinocyte cell line HaCaT. We found that the selection of the inhibitor is critical to ensure that cells do not respond by inducing counteracting activities in the context of cancer therapy, e.g., the extreme IL-8 induction mediated by MLN4924 or FasL resistance mediated by Cortisol. However, TPCA1 was qualified by this in vitro study as an excellent therapeutic mediator in HNSCC by four positive qualities: (1) proliferation was inhibited at low μM-range concentrations; (2) TNFα-induced IL-8 secretion was blocked; (3) HNSCC cells were sensitized to TNFα-induced cell death; and (4) FasL-mediated apoptosis was not disrupted.
Collapse
Affiliation(s)
| | - Roman Camillus Brands
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, D-97080 Würzburg, Germany.
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura E-35516, Egypt.
| | - Stefan Hartmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg, D-97070 Würzburg, Germany.
| | | | - Alexander Christian Kübler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.
| | - Axel Seher
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, D-97070 Würzburg, Germany.
| |
Collapse
|
37
|
Zaleska B, Karelus M, Trzewik B, Serda P. A New Way to Quinazolines, Perimidines and Dibenzo[d,f][1,3]Diazepines. JOURNAL OF CHEMICAL RESEARCH 2019. [DOI: 10.3184/030823407x203387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A synthesis of quinazolines, perimidines and dibenzo[d,f][1,3]diazepines is described. The method involves rearrangements following cyclisation of 2-anilino-2-methoxy-3-oxo-N-phenylbutanethioamides with aromatic 1,3-and 1,4-diamines.
Collapse
Affiliation(s)
- Barbara Zaleska
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Romana Ingardena 3, 30-060 Kraków, Poland
| | - Marcin Karelus
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Romana Ingardena 3, 30-060 Kraków, Poland
| | - Bartosz Trzewik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Romana Ingardena 3, 30-060 Kraków, Poland
| | - Paweł Serda
- Regional Laboratory of Physicochemical Analysis and Structural Research, ul. Romana Ingardena 3, 30-060 Kraków, Poland
| |
Collapse
|
38
|
Luo Y, Wu J, Wu Q, Li X, Wu J, Zhang J, Rong X, Rao J, Liao Y, Bin J, Huang N, Liao W. miR-577 Regulates TGF-β Induced Cancer Progression through a SDPR-Modulated Positive-Feedback Loop with ERK-NF-κB in Gastric Cancer. Mol Ther 2019; 27:1166-1182. [PMID: 30879950 PMCID: PMC6554531 DOI: 10.1016/j.ymthe.2019.02.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor β (TGF-β) drives epithelial-mesenchymal transition (EMT), playing vital roles in cancer metastasis. The crosstalk between microRNAs (miRNAs) and TGF-β are frequently observed and involved in TGF-β-induced EMT. Here, we determine that miR-577 is significantly upregulated in gastric cancer (GC). miR-577 expression is positively correlated with GC metastasis status and poor patient prognosis. Functional assays demonstrate that miR-577 promotes metastasis and chemoresistance by inducing EMT and stemness-like properties. Moreover, TGF-β promotes the expression of miR-577, and miR-577 participates TGF-β-mediated cancer metastasis. Mechanistically, TGF-β activates miR-577 via NF-κB-mediated transcription, and miR-577 enhances TGF-β signaling by targeting the serum deprivation protein response (SDPR), which directly interacts with ERK to inactivate the ERK-NF-κB pathway, hence forming a feedback loop to drive tumor metastasis. A plausible mechanism of EMT induction by the TGF-β network is elucidated. Our findings suggest that the TGF-β-miR-577-SDPR axis may be a potential prognostic marker and therapeutic target against cancer metastasis in GC.
Collapse
Affiliation(s)
- Yuhao Luo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qianying Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyin Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiani Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingwen Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingjun Rao
- Key Laboratory of New Drug Screening of Guangdong Province, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
39
|
Razumilava N, Shiota J, Mohamad Zaki NH, Ocadiz-Ruiz R, Cieslak CM, Zakharia K, Allen BL, Gores GJ, Samuelson LC, Merchant JL. Hedgehog Signaling Modulates Interleukin-33-Dependent Extrahepatic Bile Duct Cell Proliferation in Mice. Hepatol Commun 2019; 3:277-292. [PMID: 30766964 PMCID: PMC6357834 DOI: 10.1002/hep4.1295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/14/2018] [Indexed: 01/11/2023] Open
Abstract
Hedgehog (HH) signaling participates in hepatobiliary repair after injury and is activated in patients with cholangiopathies. Cholangiopathies are associated with bile duct (BD) hyperplasia, including expansion of peribiliary glands, the niche for biliary progenitor cells. The inflammation-associated cytokine interleukin (IL)-33 is also up-regulated in cholangiopathies, including cholangiocarcinoma. We hypothesized that HH signaling synergizes with IL-33 in acute inflammation-induced BD hyperplasia. We measured extrahepatic BD (EHBD) thickness and cell proliferation with and without an IL-33 challenge in wild-type mice, mice overexpressing Sonic HH (pCMV-Shh), and mice with loss of the HH pathway effector glioma-associated oncogene 1 (Gli1lacZ/lacZ ). LacZ reporter mice were used to map the expression of HH effector genes in mouse EHBDs. An EHBD organoid (BDO) system was developed to study biliary progenitor cells in vitro. EHBDs from the HH overexpressing pCMV-Shh mice showed increased epithelial cell proliferation and hyperplasia when challenged with IL-33. In Gli1lacZ/lacZ mice, we observed a decreased proliferative response to IL-33 and decreased expression of Il6. The HH ligands Shh and Indian HH (Ihh) were expressed in epithelial cells, whereas the transcriptional effectors Gli1, Gli2, and Gli3 and the HH receptor Patched1 (Ptch1) were expressed in stromal cells, as assessed by in situ hybridization and lacZ reporter mice. Although BDO cells lacked canonical HH signaling, they expressed the IL-33 receptor suppression of tumorigenicity 2. Accordingly, IL-33 treatment directly induced BDO cell proliferation in a nuclear factor κB-dependent manner. Conclusion: HH ligand overexpression enhances EHBD epithelial cell proliferation induced by IL-33. This proproliferative synergism of HH and IL-33 involves crosstalk between HH ligand-producing epithelial cells and HH-responding stromal cells.
Collapse
Affiliation(s)
| | - Junya Shiota
- Department of Internal Medicine University of Michigan Ann Arbor MI
| | | | | | | | - Kais Zakharia
- Department of Internal Medicine University of Michigan Ann Arbor MI
| | - Benjamin L Allen
- Department of Cell and Developmental Biology University of Michigan Ann Arbor MI
| | | | - Linda C Samuelson
- Department of Internal Medicine University of Michigan Ann Arbor MI
- Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| | - Juanita L Merchant
- Department of Internal Medicine University of Michigan Ann Arbor MI
- Molecular and Integrative Physiology University of Michigan Ann Arbor MI
| |
Collapse
|
40
|
Li H, Yin B, Wang S, Fu Q, Xiao B, Lǚ K, He J, Li C. RNAi screening identifies a new Toll from shrimp Litopenaeus vannamei that restricts WSSV infection through activating Dorsal to induce antimicrobial peptides. PLoS Pathog 2018; 14:e1007109. [PMID: 30256850 PMCID: PMC6175524 DOI: 10.1371/journal.ppat.1007109] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/08/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022] Open
Abstract
The function of Toll pathway defense against bacterial infection has been well established in shrimp, however how this pathway responds to viral infection is still largely unknown. In this study, we report the Toll4-Dorsal-AMPs cascade restricts the white spot syndrome virus (WSSV) infection of shrimp. A total of nine Tolls from Litopenaeus vannamei namely Toll1-9 are identified, and RNAi screening in vivo reveals the Toll4 is important for shrimp to oppose WSSV infection. Knockdown of Toll4 results in elevated viral loads and renders shrimp more susceptible to WSSV. Furthermore, Toll4 could be a one of upstream pattern recognition receptor (PRR) to detect WSSV, and thereby leading to nuclear translocation and phosphorylation of Dorsal, the known NF-κB transcription factor of the canonical Toll pathway. More importantly, silencing of Toll4 and Dorsal contributes to impaired expression of a specific set of antimicrobial peptides (AMPs) such as anti-LPS-factor (ALF) and lysozyme (LYZ) family, which exert potent anti-WSSV activity. Two AMPs of ALF1 and LYZ1 as representatives are demonstrated to have the ability to interact with several WSSV structural proteins to inhibit viral infection. Taken together, we therefore identify that the Toll4-Dorsal pathway mediates strong resistance to WSSV infection by inducing some specific AMPs. The TLR pathway mediated antiviral immune response is well identified in mammals, yet, Toll pathway governing this protection in invertebrates remains unknown. In the present study, we uncover that a shrimp Toll4 from a total of nine Tolls in L. vannamei confers resistance to WSSV thought inducing the NF-κB transcription factor Dorsal to inspire the production of some antimicrobial peptides (AMPs) with antiviral activity. The anti-LPS-factor (ALF) and lysozyme (LYZ) family are identified as the Toll4-Dorsal pathway targeted genes with the ability to interact with viral structural proteins in response to WSSV infection. These results suggest that the Toll receptor induces the expression of AMPs with antiviral activity could be a general antiviral mechanism in invertebrates and Toll pathway established antiviral defense could be conserved during evolution.
Collapse
Affiliation(s)
- Haoyang Li
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Bin Yin
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Sheng Wang
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Qihui Fu
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Bang Xiao
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Kai Lǚ
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
- * E-mail: (JH); (CL)
| | - Chaozheng Li
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
- * E-mail: (JH); (CL)
| |
Collapse
|
41
|
Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:575-590. [PMID: 30406177 PMCID: PMC6214864 DOI: 10.1016/j.trci.2018.06.014] [Citation(s) in RCA: 1404] [Impact Index Per Article: 200.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by cognitive decline and the presence of two core pathologies, amyloid β plaques and neurofibrillary tangles. Over the last decade, the presence of a sustained immune response in the brain has emerged as a third core pathology in AD. The sustained activation of the brain's resident macrophages (microglia) and other immune cells has been demonstrated to exacerbate both amyloid and tau pathology and may serve as a link in the pathogenesis of the disorder. In the following review, we provide an overview of inflammation in AD and a detailed coverage of a number of microglia-related signaling mechanisms that have been implicated in AD. Additional information on microglia signaling and a number of cytokines in AD are also reviewed. We also review the potential connection of risk factors for AD and how they may be related to inflammatory mechanisms.
Collapse
Affiliation(s)
- Jefferson W. Kinney
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Shane M. Bemiller
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew S. Murtishaw
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Amanda M. Leisgang
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Arnold M. Salazar
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
42
|
Vara D, Watt JM, Fortunato TM, Mellor H, Burgess M, Wicks K, Mace K, Reeksting S, Lubben A, Wheeler-Jones CPD, Pula G. Direct Activation of NADPH Oxidase 2 by 2-Deoxyribose-1-Phosphate Triggers Nuclear Factor Kappa B-Dependent Angiogenesis. Antioxid Redox Signal 2018; 28:110-130. [PMID: 28793782 PMCID: PMC5725637 DOI: 10.1089/ars.2016.6869] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Deoxyribose-1-phosphate (dRP) is a proangiogenic paracrine stimulus released by cancer cells, platelets, and macrophages and acting on endothelial cells. The objective of this study was to clarify how dRP stimulates angiogenic responses in human endothelial cells. RESULTS Live cell imaging, electron paramagnetic resonance, pull-down of dRP-interacting proteins, followed by immunoblotting, gene silencing of different NADPH oxidases (NOXs), and their regulatory cosubunits by small interfering RNA (siRNA) transfection, and experiments with inhibitors of the sugar transporter glucose transporter 1 (GLUT1) were utilized to demonstrate that dRP acts intracellularly by directly activating the endothelial NOX2 complex, but not NOX4. Increased reactive oxygen species generation in response to NOX2 activity leads to redox-dependent activation of the transcription factor nuclear factor kappa B (NF-κB), which, in turn, induces vascular endothelial growth factor receptor 2 (VEGFR2) upregulation. Using endothelial tube formation assays, gene silencing by siRNA, and antibody-based receptor inhibition, we demonstrate that the activation of NF-κB and VEGFR2 is necessary for the angiogenic responses elicited by dRP. The upregulation of VEGFR2 and NOX2-dependent stimulation of angiogenesis by dRP were confirmed in excisional wound and Matrigel plug vascularization assays in vivo using NOX2-/- mice. INNOVATION For the first time, we demonstrate that dRP acts intracellularly and stimulates superoxide anion generation by direct binding and activation of the NOX2 enzymatic complex. CONCLUSIONS This study describes a novel molecular mechanism underlying the proangiogenic activity of dRP, which involves the sequential activation of NOX2 and NF-κB and upregulation of VEGFR2. Antioxid. Redox Signal. 28, 110-130.
Collapse
Affiliation(s)
- Dina Vara
- 1 Institute of Biomedical and Clinical Science, University of Exeter Medical School , Exeter, United Kingdom
| | - Joanna M Watt
- 2 Department of Pharmacy and Pharmacology, University of Bath , Bath, United Kingdom
| | - Tiago M Fortunato
- 3 Department of Biomedical Engineering, Eindhoven University of Technology , Eindhoven, The Netherlands
| | - Harry Mellor
- 4 Department of Biochemistry, University of Bristol , Bristol, United Kingdom
| | - Matthew Burgess
- 5 The Healing Foundation Centre, University of Manchester , Manchester, United Kingdom
| | - Kate Wicks
- 5 The Healing Foundation Centre, University of Manchester , Manchester, United Kingdom
| | - Kimberly Mace
- 5 The Healing Foundation Centre, University of Manchester , Manchester, United Kingdom
| | - Shaun Reeksting
- 6 Mass Spectrometry Service and Chemical Characterisation and Analysis Facility, University of Bath , Bath, United Kingdom
| | - Anneke Lubben
- 6 Mass Spectrometry Service and Chemical Characterisation and Analysis Facility, University of Bath , Bath, United Kingdom
| | | | - Giordano Pula
- 1 Institute of Biomedical and Clinical Science, University of Exeter Medical School , Exeter, United Kingdom
| |
Collapse
|
43
|
Su J, Liu X, Xu C, Lu X, Wang F, Fang H, Lu A, Qiu Q, Li C, Yang T. NF-κB-dependent upregulation of (pro)renin receptor mediates high-NaCl-induced apoptosis in mouse inner medullary collecting duct cells. Am J Physiol Cell Physiol 2017; 313:C612-C620. [PMID: 29021196 DOI: 10.1152/ajpcell.00068.2017] [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] [Indexed: 01/03/2023]
Abstract
(Pro)renin receptor (PRR), a component of the renin-angiotensin system, has emerged as a new regulator of collecting duct function. The present study was designed to investigate the role of PRR in high salt-induced apoptosis in cultured mouse inner medullary collecting duct cells, mIMCD-K2 cells. Exposure to high NaCl at 550 mosM/kgH2O increased PRR protein abundance, as did exposure to mannitol, sodium gluconate, or choline chloride. This was accompanied by upregulation of the abundance of phosphorylated NF-κB p65 protein. NF-κB inhibition with QNZ, caffeic acid phenethyl ester, or small interfering RNA (siRNA)-mediated silencing of NF-κB p65 attenuated high-NaCl-induced PRR upregulation. Exposure to high salt for 24 h induced apoptosis, as assessed by immunoblotting and immunocytochemistry analysis of cleaved caspase-3 and flow cytometry analysis of the number of apoptotic cells. High-NaCl-induced apoptosis was attenuated by a PRR decoy inhibitor, PRO20, or siRNA-mediated silencing of NF-κB p65. These results show that induction of PRR expression by exposure to high NaCl occurs through activation of NF-κB, thus contributing to cell apoptosis.
Collapse
Affiliation(s)
- Jiahui Su
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Xiyang Liu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Chuanming Xu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Xiaohan Lu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China.,Department of Internal Medicine, University of Utah, and Veterans Affairs Medical Center , Salt Lake City, Utah
| | - Fei Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China.,Department of Internal Medicine, University of Utah, and Veterans Affairs Medical Center , Salt Lake City, Utah
| | - Hui Fang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Aihua Lu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Qixiang Qiu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Tianxin Yang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China.,Department of Internal Medicine, University of Utah, and Veterans Affairs Medical Center , Salt Lake City, Utah
| |
Collapse
|
44
|
Synergistic Toxicity of Polyglutamine-Expanded TATA-Binding Protein in Glia and Neuronal Cells: Therapeutic Implications for Spinocerebellar Ataxia 17. J Neurosci 2017; 37:9101-9115. [PMID: 28821675 DOI: 10.1523/jneurosci.0111-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/09/2017] [Accepted: 07/24/2017] [Indexed: 01/12/2023] Open
Abstract
Spinocerebellar ataxia 17 (SCA17) is caused by polyglutamine (polyQ) repeat expansion in the TATA-binding protein (TBP) and is among a family of neurodegenerative diseases in which polyQ expansion leads to preferential neuronal loss in the brain. Although previous studies have demonstrated that expression of polyQ-expanded proteins in glial cells can cause neuronal injury via noncell-autonomous mechanisms, these studies investigated animal models that overexpress transgenic mutant proteins. Since glial cells are particularly reactive to overexpressed mutant proteins, it is important to investigate the in vivo role of glial dysfunction in neurodegeneration when mutant polyQ proteins are endogenously expressed. In the current study, we generated two conditional TBP-105Q knock-in mouse models that specifically express mutant TBP at the endogenous level in neurons or in astrocytes. We found that mutant TBP expression in neuronal cells or astrocytes alone only caused mild neurodegeneration, whereas severe neuronal toxicity requires the expression of mutant TBP in both neuronal and glial cells. Coculture of neurons and astrocytes further validated that mutant TBP in astrocytes promoted neuronal injury. We identified activated inflammatory signaling pathways in mutant TBP-expressing astrocytes, and blocking nuclear factor κB (NF-κB) signaling in astrocytes ameliorated neurodegeneration. Our results indicate that the synergistic toxicity of mutant TBP in neuronal and glial cells plays a critical role in SCA17 pathogenesis and that targeting glial inflammation could be a potential therapeutic approach for SCA17 treatment.SIGNIFICANCE STATEMENT Mutant TBP with polyglutamine expansion preferentially affects neuronal viability in SCA17 patients. Whether glia, the cells that support and protect neurons, contribute to neurodegeneration in SCA17 remains mostly unexplored. In this study, we provide both in vivo and in vitro evidence arguing that endogenous expression of mutant TBP in neurons and glia synergistically impacts neuronal survival. Hyperactivated inflammatory signaling pathways, particularly the NF-κB pathway, underlie glia-mediated neurotoxicity. Moreover, blocking NF-κB activity with small chemical inhibitors alleviated such neurotoxicity. Our study establishes glial dysfunction as an important component of SCA17 pathogenesis and suggests targeting glial inflammation as a potential therapeutic approach for SCA17 treatment.
Collapse
|
45
|
Hamed MM, Darwish SS, Herrmann J, Abadi AH, Engel M. First Bispecific Inhibitors of the Epidermal Growth Factor Receptor Kinase and the NF-κB Activity As Novel Anticancer Agents. J Med Chem 2017; 60:2853-2868. [PMID: 28291344 DOI: 10.1021/acs.jmedchem.6b01774] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The activation of the NF-κB transcription factor is a major adaptive response induced upon treatment with EGFR kinase inhibitors, leading to the emergence of resistance in nonsmall cell lung cancer and other tumor types. To suppress this survival mechanism, we developed new thiourea quinazoline derivatives that are dual inhibitors of both EGFR kinase and the NF-κB activity. Optimization of the hit compound, identified in a NF-κB reporter gene assay, led to compound 9b, exhibiting a cellular IC50 for NF-κB inhibition of 0.3 μM while retaining a potent EGFR kinase inhibition (IC50 = 60 nM). The dual inhibitors showed a higher potency than gefitinib to inhibit cell growth of EGFR-overexpressing tumor cell lines in vitro and in a xenograft model in vivo, while no signs of toxicity were observed. An investigation of the molecular mechanism of NF-κB suppression revealed that the dual inhibitors depleted the transcriptional coactivator CREB-binding protein from the NF-κB complex in the nucleus.
Collapse
Affiliation(s)
- Mostafa M Hamed
- Pharmaceutical and Medicinal Chemistry, Campus C2.3, and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarland University , Campus E8.1, D-66123 Saarbrücken, Germany.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Sarah S Darwish
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, D-66123 Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, D-66123 Saarbrücken, Germany
| |
Collapse
|
46
|
A novel quinazoline-based analog induces G2/M cell cycle arrest and apoptosis in human A549 lung cancer cells via a ROS-dependent mechanism. Biochem Biophys Res Commun 2017; 486:314-320. [PMID: 28302490 DOI: 10.1016/j.bbrc.2017.03.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/11/2017] [Indexed: 12/16/2022]
Abstract
6-amino-4-(4-phenoxyphenylethylamino)quinazoline (QNZ) is an excellent quinazoline-containing NF-κB inhibitor also acting as a novel anticancer agent. Considering both the medicinal significance of quinazoline scaffold and the tunable functionality of Michael acceptor-centric pharmacophores in the electrophilicity-based prooxidant strategy, we designed a novel QNZ-inspired electrophilic molecule QNZ-A by introducing a Michael acceptor unit at position-6 of quinazoline ring in QNZ. Our results identified QNZ-A as a promising selective cytotoxic agent against A549 cells. QNZ-A, by virtue of its Michael acceptor unit, induced reactive oxygen species (ROS) accumulation associated with collapse of the redox buffering system in A549 cells. This caused up-regulation of p53-inducible p21 and down-regulation of redox sensitive Cdc25C along with Cyclin B1/Cdk1, leading to a G2/M cell cycle arrest and final cell apoptosis. By contrast, QNZ-B, a reduction product of QNZ-A lacking the Michael acceptor unit failed to induce ROS generation and all these cell cycle-related events. In conclusion, this work provided a successful example of designing QNZ-directed anticancer agent by a ROS-promoting strategy and identified QNZ-A as a selective anticancer agent against A549 cells through G2/M cell cycle arrest and apoptosis via a ROS-dependent mechanism.
Collapse
|
47
|
Krishnathas R, Bonke E, Dröse S, Zickermann V, Nasiri HR. Identification of 4- N-[2-(4-phenoxyphenyl)ethyl]quinazoline-4,6-diamine as a novel, highly potent and specific inhibitor of mitochondrial complex I. MEDCHEMCOMM 2017; 8:657-661. [PMID: 30108783 DOI: 10.1039/c6md00655h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022]
Abstract
By probing the quinone substrate binding site of mitochondrial complex I with a focused set of quinazoline-based compounds, we identified substitution patterns as being critical for the observed inhibition. The structure activity relationship study also resulted in the discovery of the quinazoline 4-N-[2-(4-phenoxyphenyl)ethyl]quinazoline-4,6-diamine (EVP4593) as a highly potent inhibitor of the multisubunit membrane protein. EVP4593 specifically and effectively reduces the mitochondrial complex I-dependent respiration with no effect on the respiratory chain complexes II-IV. Similar to established Q-site inhibitors, EVP4593 elicits the release of reactive oxygen species at the flavin site of mitochondrial complex I. Recently, EVP4593 was nominated as a lead compound for the treatment of Huntingtons disease. Our results challenge the postulated primary mode-of-action of EVP4593 as an inhibitor of NF-κB pathway activation and/or store-operated calcium influx.
Collapse
Affiliation(s)
- Robin Krishnathas
- Johann Wolfgang Goethe-University Frankfurt , Max-von-Laue-Straße 7 , D-60438 Frankfurt am Main , Germany .
| | - Erik Bonke
- Department of Anaesthesiology , Intensive-Care Medicine and Pain Therapy , University Hospital Frankfurt , 60590 Frankfurt am Main , Germany
| | - Stefan Dröse
- Department of Anaesthesiology , Intensive-Care Medicine and Pain Therapy , University Hospital Frankfurt , 60590 Frankfurt am Main , Germany
| | - Volker Zickermann
- Structural Bioenergetics Group , Institute of Biochemistry II , Medical School , Goethe-University , 60438 Frankfurt am Main , Germany.,Cluster of Excellence Frankfurt "Macromolecular Complexes," , Goethe-University , 60438 Frankfurt am Main , Germany
| | - Hamid R Nasiri
- Johann Wolfgang Goethe-University Frankfurt , Max-von-Laue-Straße 7 , D-60438 Frankfurt am Main , Germany .
| |
Collapse
|
48
|
Singh AK, Awasthi D, Dubey M, Nagarkoti S, Kumar A, Chandra T, Barthwal MK, Tripathi AK, Dikshit M. High oxidative stress adversely affects NFκB mediated induction of inducible nitric oxide synthase in human neutrophils: Implications in chronic myeloid leukemia. Nitric Oxide 2016; 58:28-41. [PMID: 27264783 DOI: 10.1016/j.niox.2016.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
Increasing evidence support bimodal action of nitric oxide (NO) both as a promoter and as an impeder of oxygen free radicals in neutrophils (PMNs), however impact of high oxidative stress on NO generation is less explored. In the present study, we comprehensively investigated the effect of high oxidative stress on inducible nitric oxide synthase (iNOS) expression and NO generation in human PMNs. Our findings suggest that PMA or diamide induced oxidative stress in PMNs from healthy volunteers, and high endogenous ROS in PMNs of chronic myeloid leukemia (CML) patients attenuate basal as well as LPS/cytokines induced NO generation and iNOS expression in human PMNs. Mechanistically, we found that under high oxidative stress condition, S-glutathionylation of NFκB (p50 and p65 subunits) severely limits iNOS expression due to its reduced binding to iNOS promoter, which was reversed in presence of DTT. Furthermore, by using pharmacological inhibitors, scavengers and molecular approaches, we identified that enhanced ROS generation via NOX2 and mitochondria, reduced Grx1/2 expression and GSH level associated with NFκB S-glutathionylation in PMNs from CML patients. Altogether data obtained suggest that oxidative status act as an important regulator of NO generation/iNOS expression, and under enhanced oxidative stress condition, NOX2-mtROS-NFκB S-glutathionylation is a feed forward loop, which attenuate NO generation and iNOS expression in human PMNs.
Collapse
Affiliation(s)
| | - Deepika Awasthi
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Megha Dubey
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sheela Nagarkoti
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ashutosh Kumar
- Department of Pathology, King George's Medical University, Lucknow, India
| | - Tulika Chandra
- Department of Transfusion Medicine, King George's Medical University, Lucknow, India
| | | | - Anil Kumar Tripathi
- Department of Clinical Haematology & Medical Oncology, King George's Medical University, Lucknow, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
49
|
Progatzky F, Cook HT, Lamb JR, Bugeon L, Dallman MJ. Mucosal inflammation at the respiratory interface: a zebrafish model. Am J Physiol Lung Cell Mol Physiol 2016; 310:L551-61. [PMID: 26719149 PMCID: PMC4796261 DOI: 10.1152/ajplung.00323.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022] Open
Abstract
Inflammatory diseases of the respiratory system such as asthma and chronic obstructive pulmonary disease are increasing globally and remain poorly understood conditions. Although attention has long focused on the activation of type 1 and type 2 helper T cells of the adaptive immune system in these diseases, it is becoming increasingly apparent that there is also a need to understand the contributions and interactions between innate immune cells and the epithelial lining of the respiratory system. Cigarette smoke predisposes the respiratory tissue to a higher incidence of inflammatory disease, and here we have used zebrafish gills as a model to study the effect of cigarette smoke on the respiratory epithelium. Zebrafish gills fulfill the same gas-exchange function as the mammalian airways and have a similar structure. Exposure to cigarette smoke extracts resulted in an increase in transcripts of the proinflammatory cytokines TNF-α, IL-1β, and MMP9 in the gill tissue, which was at least in part mediated via NF-κB activation. Longer term exposure of fish for 6 wk to cigarette smoke extract resulted in marked structural changes to the gills with lamellar fusion and mucus cell formation, while signs of inflammation or fibrosis were absent. This shows, for the first time, that zebrafish gills are a relevant model for studying the effect of inflammatory stimuli on a respiratory epithelium, since they mimic the immunopathology involved in respiratory inflammatory diseases of humans.
Collapse
Affiliation(s)
- Fränze Progatzky
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, United Kingdom; and
| | - H Terence Cook
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jonathan R Lamb
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, United Kingdom; and
| | - Laurence Bugeon
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, United Kingdom; and
| | - Margaret J Dallman
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, United Kingdom; and
| |
Collapse
|
50
|
Böhringer M, Pohlers S, Schulze S, Albrecht-Eckardt D, Piegsa J, Weber M, Martin R, Hünniger K, Linde J, Guthke R, Kurzai O. Candida albicans infection leads to barrier breakdown and a MAPK/NF-κB mediated stress response in the intestinal epithelial cell line C2BBe1. Cell Microbiol 2016; 18:889-904. [PMID: 26752615 DOI: 10.1111/cmi.12566] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 12/04/2015] [Accepted: 12/20/2015] [Indexed: 12/15/2022]
Abstract
Intestinal epithelial cells (IEC) form a tight barrier to the gut lumen. Paracellular permeability of the intestinal barrier is regulated by tight junction proteins and can be modulated by microorganisms and other stimuli. The polymorphic fungus Candida albicans, a frequent commensal of the human mucosa, has the capacity of traversing this barrier and establishing systemic disease within the host. Infection of polarized C2BBe1 IEC with wild-type C. albicans led to a transient increase of transepithelial electric resistance (TEER) before subsequent barrier disruption, accompanied by a strong decline of junctional protein levels and substantial, but considerably delayed cytotoxicity. Time-resolved microarray-based transcriptome analysis of C. albicans challenged IEC revealed a prominent role of NF-κB and MAPK signalling pathways in the response to infection. Hence, we inferred a gene regulatory network based on differentially expressed NF-κB and MAPK pathway components and their predicted transcriptional targets. The network model predicted activation of GDF15 by NF-κB was experimentally validated. Furthermore, inhibition of NF-κB activation in C. albicans infected C2BBe1 cells led to enhanced cytotoxicity in the epithelial cells. Taken together our study identifies NF-κB activation as an important protective signalling pathway in the response of epithelial cells to C. albicans.
Collapse
Affiliation(s)
- Michael Böhringer
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Susann Pohlers
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Sylvie Schulze
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | | | - Judith Piegsa
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Michael Weber
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Ronny Martin
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Kerstin Hünniger
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Jörg Linde
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Reinhard Guthke
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Oliver Kurzai
- Septomics Research Centre, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany.,German National Reference Center for Invasive Fungal Infections, Hans Knöll Institute, Jena, Germany
| |
Collapse
|