1
|
Vymazal O, Papatheodorou I, Andrejčinová I, Bosáková V, Vascelli G, Bendíčková K, Zelante T, Hortová-Kohoutková M, Frič J. Calcineurin-NFAT signaling controls neutrophils' ability of chemoattraction upon fungal infection. J Leukoc Biol 2024; 116:816-829. [PMID: 38648505 DOI: 10.1093/jleuko/qiae091] [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: 12/01/2023] [Revised: 03/03/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Calcineurin-nuclear factor of activated T cells (CN-NFAT) inhibitors are widely clinically used drugs for immunosuppression, but besides their required T cell response inhibition, they also undesirably affect innate immune cells. Disruption of innate immune cell function can explain the observed susceptibility of CN-NFAT inhibitor-treated patients to opportunistic fungal infections. Neutrophils play an essential role in innate immunity as a defense against pathogens; however, the effect of CN-NFAT inhibitors on neutrophil function was poorly described. Thus, we tested the response of human neutrophils to opportunistic fungal pathogens, namely Candida albicans and Aspergillus fumigatus, in the presence of CN-NFAT inhibitors. Here, we report that the NFAT pathway members were expressed in neutrophils and mediated part of the neutrophil response to pathogens. Upon pathogen exposure, neutrophils underwent profound transcriptomic changes with subsequent production of effector molecules. Importantly, genes and proteins involved in the regulation of the immune response and chemotaxis, including the chemokines CCL2, CCL3, and CCL4 were significantly upregulated. The presence of CN-NFAT inhibitors attenuated the expression of these chemokines and impaired the ability of neutrophils to chemoattract other immune cells. Our results amend knowledge about the impact of CN-NFAT inhibition in human neutrophils.
Collapse
Affiliation(s)
- Ondrej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ioanna Papatheodorou
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Gianluca Vascelli
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Teresa Zelante
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Marcela Hortová-Kohoutková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
- Institute of Hematology and Blood Transfusion, U Nemocnice 2094/1, Prague 2, 128 00, Czech Republic
| |
Collapse
|
2
|
Pernaute OS. Voclosporin is on the table, let's taste it. Rheumatology (Oxford) 2024; 63:1762-1765. [PMID: 38224548 DOI: 10.1093/rheumatology/keae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/01/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024] Open
Affiliation(s)
- Olga Sánchez Pernaute
- Unit for Autoimmune Diseases, Rheumatology Department, Fundación Jiménez Díaz University Hospital and Health Research Institute (IIS-FJD), Madrid, Spain
| |
Collapse
|
3
|
Qu Y, Wang S, Jiang H, Wang Q, Liao Y, Qiu X, Tan L, Song C, Ding C, Sun Y, Yang Z. The Ca 2+-dependent phosphatase calcineurin dephosphorylates TBK1 to suppress antiviral innate immunity. J Virol 2024; 98:e0001624. [PMID: 38563732 PMCID: PMC11092360 DOI: 10.1128/jvi.00016-24] [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: 02/06/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Tumor necrosis factor receptor-associated factor family member-associated NF-κB activator-binding kinase 1 (TBK1) plays a key role in the induction of the type 1 interferon (IFN-I) response, which is an important component of innate antiviral defense. Viruses target calcium (Ca2+) signaling networks, which participate in the regulation of the viral life cycle, as well as mediate the host antiviral response. Although many studies have focused on the role of Ca2+ signaling in the regulation of IFN-I, the relationship between Ca2+ and TBK1 in different infection models requires further elucidation. Here, we examined the effects of the Newcastle disease virus (NDV)-induced increase in intracellular Ca2+ levels on the suppression of host antiviral responses. We demonstrated that intracellular Ca2+ increased significantly during NDV infection, leading to impaired IFN-I production and antiviral immunity through the activation of calcineurin (CaN). Depletion of Ca²+ was found to lead to a significant increase in virus-induced IFN-I production resulting in the inhibition of viral replication. Mechanistically, the accumulation of Ca2+ in response to viral infection increases the phosphatase activity of CaN, which in turn dephosphorylates and inactivates TBK1 in a Ca2+-dependent manner. Furthermore, the inhibition of CaN on viral replication was counteracted in TBK1 knockout cells. Together, our data demonstrate that NDV hijacks Ca2+ signaling networks to negatively regulate innate immunity via the CaN-TBK1 signaling axis. Thus, our findings not only identify the mechanism by which viruses exploit Ca2+ signaling to evade the host antiviral response but also, more importantly, highlight the potential role of Ca2+ homeostasis in the viral innate immune response.IMPORTANCEViral infections disrupt intracellular Ca2+ homeostasis, which affects the regulation of various host processes to create conditions that are conducive for their own proliferation, including the host immune response. The mechanism by which viruses trigger TBK1 activation and IFN-I induction through viral pathogen-associated molecular patterns has been well defined. However, the effects of virus-mediated Ca2+ imbalance on the IFN-I pathway requires further elucidation, especially with respect to TBK1 activation. Herein, we report that NDV infection causes an increase in intracellular free Ca2+ that leads to activation of the serine/threonine phosphatase CaN, which subsequently dephosphorylates TBK1 and negatively regulates IFN-I production. Furthermore, depletion of Ca2+ or inhibition of CaN activity exerts antiviral effects by promoting the production of IFN-I and inhibiting viral replication. Thus, our results reveal the potential role of Ca2+ in the innate immune response to viruses and provide a theoretical reference for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Yang Qu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Siyuan Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hui Jiang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Qingyi Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
4
|
Lu Y, Liu S, Wang P, Guo X, Qin Z, Hou H, Tao T. A novel microglia-targeting strategy based on nanoparticle-mediated delivery of miR-26a-5p for long-lasting analgesia in chronic pain. J Nanobiotechnology 2024; 22:128. [PMID: 38519978 PMCID: PMC10960380 DOI: 10.1186/s12951-024-02420-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
Accumulating evidence supports the notion that microglia play versatile roles in different chronic pain conditions. However, therapeutic strategies of chronic pain by targeting microglia remain largely overlooked. This study seeks to develop a miRNA-loaded nano-delivery system by targeting microglia, which could provide a decent and long-lasting analgesia for chronic pain. Surface aminated mesoporous silica nanoparticles were adopted to load miR-26a-5p, a potent analgesic miRNA, by electrostatic adsorption, which can avoid miR-26a-5p is rapidly released and degraded. Then, targeting peptide MG1 was modified on the surface of aminated mesoporous silica particles for microglia targeting. In peripheral nerve injury induced neuropathic pain model, a satisfactory anti-allodynia effect with about 6 weeks pain-relief duration were achieved through targeting microglia strategy, which decreased microglia activation and inflammation by Wnt5a, a non-canonical Wnt pathway. In inflammatory pain and chemotherapy induced peripheral neuropathic pain, microglia targeting strategy also exhibited more efficient analgesia and longer pain-relief duration than others. Overall, we developed a microglia-targeting nano-delivery system, which facilitates precisely miR-26a-5p delivery to enhance analgesic effect and duration for several chronic pain conditions.
Collapse
Affiliation(s)
- Yitian Lu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Peng Wang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiangna Guo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Tao Tao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- Department of Anesthesiology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
5
|
Liu Y, Zhang L, Wang L, Tang X, Wan S, Huang Q, Ran M, Shen H, Yang Y, Chiampanichayakul S, Tima S, Anuchapreeda S, Wu J. Targeting CD38/ ADP-ribosyl cyclase as a novel therapeutic strategy for identification of three potent agonists for leukopenia treatment. Pharmacol Res 2024; 200:107068. [PMID: 38232908 DOI: 10.1016/j.phrs.2024.107068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Leukopenia is the most common side effect of chemotherapy and radiotherapy. It potentially deteriorates into a life-threatening complication in cancer patients. Despite several agents being approved for clinical administration, there are still high incidences of pathogen-related disease due to a lack of functional immune cells. ADP-ribosyl cyclase of CD38 displays a regulatory effect on leukopoiesis and the immune system. To explore whether the ADP-ribosyl cyclase was a potential therapeutic target of leukopenia. We established a drug screening model based on an ADP-ribosyl cyclase-based pharmacophore generation algorithm and discovered three novel ADP-ribosyl cyclase agonists: ziyuglycoside II (ZGSII), brevifolincarboxylic acid (BA), and 3,4-dihydroxy-5-methoxybenzoic acid (DMA). Then, in vitro experiments demonstrated that these three natural compounds significantly promoted myeloid differentiation and antibacterial activity in NB4 cells. In vivo, experiments confirmed that the compounds also stimulated the recovery of leukocytes in irradiation-induced mice and zebrafish. The mechanism was investigated by network pharmacology, and the top 12 biological processes and the top 20 signaling pathways were obtained by intersecting target genes among ZGSII, BA, DMA, and leukopenia. The potential signaling molecules involved were further explored through experiments. Finally, the ADP-ribosyl cyclase agonists (ZGSII, BA, and DMA) has been found to regenerate microbicidal myeloid cells to effectively ameliorate leukopenia-associated infection by activating CD38/ADP-ribosyl cyclase-Ca2+-NFAT. In summary, this study constructs a drug screening model to discover active compounds against leukopenia, reveals the critical roles of ADP-ribosyl cyclase in promoting myeloid differentiation and the immune response, and provides a promising strategy for the treatment of radiation-induced leukopenia.
Collapse
Affiliation(s)
- Yuanzhi Liu
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Linwei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoqin Tang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengli Wan
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qianqian Huang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Mei Ran
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hongping Shen
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Yang
- Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Sawitree Chiampanichayakul
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Singkome Tima
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
6
|
Zmonarski SC, Banasik M, Żabińska M, Gołębiowski T, Zmonarska JM, Krajewska M. Toll-Like Receptor 3 mRNA Expression of Peripheral Blood Mononuclear Cells Identifies Kidney Recipients with Potential for Improved Graft Performance. Ann Transplant 2023; 28:e941266. [PMID: 38013407 PMCID: PMC10693178 DOI: 10.12659/aot.941266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 09/12/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Toll-like receptor 3 expression is detected both on the cell membrane and in endosomes of peripheral blood mononuclear cells (PBMC). Our goal in this study was to determine to what extent a single, baseline measurement of non-stimulated PBMC TLR3-mRNA can be related to baseline GFR (b-GFR) and post-follow-up-GFR (F-up-GFR) of a kidney transplant (KT) and baseline immunosuppression. MATERIAL AND METHODS In non-stimulated PBMC we investigated averaged mRNA expression of Toll-like receptor 3. A total of 133 patients were enrolled; the median of months after KT surgery was 11.4, with median F-up at 21.3 months. A favorable course (FCF) was determined if F-up-eGFR improved. An unfavorable course (UCF) was determined if F-up-eGFR was lower at the end of the observation. RESULTS The highest TLR3-mRNA expression was at b-GFR grade 3b; it was moderately higher at b-GFR grade 3a, and marginally higher at b-GFR grades 1+2. Most of the FCF group had b-GFR grade 3b, less frequent obesity, more effective immunosuppression, and much higher TLR3-mRNA (59% of cases were in the high-TLR3 area). Both delayed graft function (DGF) and TLR3-mRNA range below the median for the entire KT cohort (low-TLR3 area) had a negative association with b-GFR. The UCF group had more frequent DGFs and obesity, less effective immunosuppression, and lower TLR3-mRNA. CONCLUSIONS In patients with GFR grade 3, high levels of TLR3-mRNA are associated with improved graft efficacy. In patients with impaired graft function, low TLR3- mRNA expression reduces the likelihood of improved renal graft function.
Collapse
Affiliation(s)
- Sławomir C. Zmonarski
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Wrocław, Poland
| | - Mirosław Banasik
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Wrocław, Poland
| | - Marcelina Żabińska
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Wrocław, Poland
| | - Tomasz Gołębiowski
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Wrocław, Poland
| | | | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
7
|
Hadpech S, Chaiyarit S, Thongboonkerd V. Calcineurin B inhibits calcium oxalate crystallization, growth and aggregation via its high calcium-affinity property. Comput Struct Biotechnol J 2023; 21:3854-3864. [PMID: 37593722 PMCID: PMC10427926 DOI: 10.1016/j.csbj.2023.07.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 08/19/2023] Open
Abstract
Calcineurin inhibitors (CNIs) are widely used in organ transplantation to suppress immunity and prevent allograft rejection. However, some transplant patients receiving CNIs have hypocitraturia, hyperoxaluria and kidney stone with unclear mechanism. We hypothesized that CNIs suppress activities of urinary calcineurin, which may serve as the stone inhibitor. This study aimed to investigate effects of calcineurin B (CNB) on calcium oxalate monohydrate (COM) stone formation. Sequence and structural analyses revealed that CNB contained four EF-hand (Ca2+-binding) domains, which are known to regulate Ca2+ homeostasis and likely to affect COM crystals. Various crystal assays revealed that CNB dramatically inhibited COM crystallization, crystal growth and crystal aggregation. At an equal amount, degrees of its inhibition against crystallization and crystal growth were slightly inferior to total urinary proteins (TUPs) from healthy subjects that are known to strongly inhibit COM stone formation. Surprisingly, its inhibitory effect against crystal aggregation was slightly superior to TUPs. While TUPs dramatically inhibited crystal-cell adhesion, CNB had no effect on this process. Ca2+-affinity assay revealed that CNB strongly bound Ca2+ at a comparable degree as of TUPs. These findings indicate that CNB serves as a novel inhibitor of COM crystallization, growth and aggregation via its high Ca2+-affinity property.
Collapse
Affiliation(s)
- Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sakdithep Chaiyarit
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Vázquez-Blomquist D, Ramón AC, Rosales M, Pérez GV, Rosales A, Palenzuela D, Perera Y, Perea SE. Gene expression profiling unveils the temporal dynamics of CIGB-300-regulated transcriptome in AML cell lines. BMC Genomics 2023; 24:373. [PMID: 37400761 DOI: 10.1186/s12864-023-09472-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Protein kinase CK2 activity is implicated in the pathogenesis of various hematological malignancies like Acute Myeloid Leukemia (AML) that remains challenging concerning treatment. This kinase has emerged as an attractive molecular target in therapeutic. Antitumoral peptide CIGB-300 blocks CK2 phospho-acceptor sites on their substrates but it also binds to CK2α catalytic subunit. Previous proteomic and phosphoproteomic experiments showed molecular and cellular processes with relevance for the peptide action in diverse AML backgrounds but earlier transcriptional level events might also support the CIGB-300 anti-leukemic effect. Here we used a Clariom S HT assay for gene expression profiling to study the molecular events supporting the anti-leukemic effect of CIGB-300 peptide on HL-60 and OCI-AML3 cell lines. RESULTS We found 183 and 802 genes appeared significantly modulated in HL-60 cells at 30 min and 3 h of incubation with CIGB-300 for p < 0.01 and FC > = │1.5│, respectively; while 221 and 332 genes appeared modulated in OCI-AML3 cells. Importantly, functional enrichment analysis evidenced that genes and transcription factors related to apoptosis, cell cycle, leukocyte differentiation, signaling by cytokines/interleukins, and NF-kB, TNF signaling pathways were significantly represented in AML cells transcriptomic profiles. The influence of CIGB-300 on these biological processes and pathways is dependent on the cellular background, in the first place, and treatment duration. Of note, the impact of the peptide on NF-kB signaling was corroborated by the quantification of selected NF-kB target genes, as well as the measurement of p50 binding activity and soluble TNF-α induction. Quantification of CSF1/M-CSF and CDKN1A/P21 by qPCR supports peptide effects on differentiation and cell cycle. CONCLUSIONS We explored for the first time the temporal dynamics of the gene expression profile regulated by CIGB-300 which, along with the antiproliferative mechanism, can stimulate immune responses by increasing immunomodulatory cytokines. We provided fresh molecular clues concerning the antiproliferative effect of CIGB-300 in two relevant AML backgrounds.
Collapse
Affiliation(s)
- Dania Vázquez-Blomquist
- Pharmacogenomic Group, Department of System Biology, Biomedical Research Division, Center for Genetic Engineering & Biotechnology (CIGB), 10600, Havana, Cuba.
| | - Ailyn C Ramón
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, CIGB, 10600, Havana, Cuba
| | - Mauro Rosales
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, CIGB, 10600, Havana, Cuba
- Department of Animal and Human Biology, Faculty of Biology, University of Havana (UH), 10400, Havana, Cuba
| | - George V Pérez
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, CIGB, 10600, Havana, Cuba
| | - Ailenis Rosales
- Department of Animal and Human Biology, Faculty of Biology, University of Havana (UH), 10400, Havana, Cuba
| | - Daniel Palenzuela
- Pharmacogenomic Group, Department of System Biology, Biomedical Research Division, Center for Genetic Engineering & Biotechnology (CIGB), 10600, Havana, Cuba
| | - Yasser Perera
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, CIGB, 10600, Havana, Cuba.
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Hunan Province, Yongzhou Zhong Gu Biotechnology Co., Ltd, Lengshuitan District, Yongzhou City, 425000, China.
| | - Silvio E Perea
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, CIGB, 10600, Havana, Cuba.
| |
Collapse
|
9
|
Borges VDF, Galant LS, Kanashiro A, Castanheira FVES, Monteiro VVS, Duarte DÂ, Rodrigues FC, Silva CMDS, Schneider AH, Cebinelli GCM, de Lima MHF, Viola JPDB, Cunha TM, da Costa Neto CM, Alves-Filho JCF, Pupo AS, Cunha FDQ. FK506 impairs neutrophil migration that results in increased polymicrobial sepsis susceptibility. Inflamm Res 2023; 72:203-215. [PMID: 36401631 DOI: 10.1007/s00011-022-01669-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the effects of FK506 on experimental sepsis immunopathology. It investigated the effect of FK506 on leukocyte recruitment to the site of infection, systemic cytokine production, and organ injury in mice with sepsis. METHODS Using a murine cecal ligation and puncture (CLP) peritonitis model, the experiments were performed with wild-type (WT) mice and mice deficient in the gene Nfat1 (Nfat1-/-) in the C57BL/6 background. Animals were treated with 2.0 mg/kg of FK506, subcutaneously, 1 h before the sepsis model, twice a day (12 h/12 h). The number of bacteria colony forming units (CFU) was manually counted. The number of neutrophils in the lungs was estimated by the myeloperoxidase (MPO) assay. The expression of CXCR2 in neutrophils was determined using flow cytometry analysis. The expression of inflammatory cytokines in macrophage was determined using ELISA. The direct effect of FK506 on CXCR2 internalization was evaluated using HEK-293T cells after CXCL2 stimulation by the BRET method. RESULTS FK506 treatment potentiated the failure of neutrophil migration into the peritoneal cavity, resulting in bacteremia and an exacerbated systemic inflammatory response, which led to higher organ damage and mortality rates. Failed neutrophil migration was associated with elevated CXCL2 chemokine plasma levels and lower expression of the CXCR2 receptor on circulating neutrophils compared with non-treated CLP-induced septic mice. FK506 did not directly affect CXCL2-induced CXCR2 internalization by transfected HEK-293 cells or mice neutrophils, despite increasing CXCL2 release by LPS-treated macrophages. Finally, the CLP-induced response of Nfat1-/- mice was similar to those observed in the Nfat1+/+ genotype, suggesting that the FK506 effect is not dependent on the NFAT1 pathway. CONCLUSION Our data indicate that the increased susceptibility to infection of FK506-treated mice is associated with failed neutrophil migration due to the reduced membrane availability of CXCR2 receptors in response to exacerbated levels of circulating CXCL2.
Collapse
Affiliation(s)
- Vanessa de Fátima Borges
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Leticia Selinger Galant
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Kanashiro
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Vargas E Silva Castanheira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Valter Vinícius Silva Monteiro
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Diego Ângelo Duarte
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Filipe Camargo Rodrigues
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Meirelles de Souza Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ayda Henriques Schneider
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme Cesar Martelossi Cebinelli
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mikhael Haruo Fernandes de Lima
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Claudio Miguel da Costa Neto
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José Carlos Farias Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - André Sampaio Pupo
- Department of Biophysics and Pharmacology, Institute of Biosciences, University of São Paulo State (UNESP), Botucatu, São Paulo, Brazil
| | - Fernando de Queiroz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
10
|
Tubiana J, Adriana-Lifshits L, Nissan M, Gabay M, Sher I, Sova M, Wolfson HJ, Gal M. Funneling modulatory peptide design with generative models: Discovery and characterization of disruptors of calcineurin protein-protein interactions. PLoS Comput Biol 2023; 19:e1010874. [PMID: 36730443 PMCID: PMC9928118 DOI: 10.1371/journal.pcbi.1010874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/14/2023] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Design of peptide binders is an attractive strategy for targeting "undruggable" protein-protein interfaces. Current design protocols rely on the extraction of an initial sequence from one known protein interactor of the target protein, followed by in-silico or in-vitro mutagenesis-based optimization of its binding affinity. Wet lab protocols can explore only a minor portion of the vast sequence space and cannot efficiently screen for other desirable properties such as high specificity and low toxicity, while in-silico design requires intensive computational resources and often relies on simplified binding models. Yet, for a multivalent protein target, dozens to hundreds of natural protein partners already exist in the cellular environment. Here, we describe a peptide design protocol that harnesses this diversity via a machine learning generative model. After identifying putative natural binding fragments by literature and homology search, a compositional Restricted Boltzmann Machine is trained and sampled to yield hundreds of diverse candidate peptides. The latter are further filtered via flexible molecular docking and an in-vitro microchip-based binding assay. We validate and test our protocol on calcineurin, a calcium-dependent protein phosphatase involved in various cellular pathways in health and disease. In a single screening round, we identified multiple 16-length peptides with up to six mutations from their closest natural sequence that successfully interfere with the binding of calcineurin to its substrates. In summary, integrating protein interaction and sequence databases, generative modeling, molecular docking and interaction assays enables the discovery of novel protein-protein interaction modulators.
Collapse
Affiliation(s)
- Jérôme Tubiana
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Lucia Adriana-Lifshits
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Nissan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Matan Gabay
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Sher
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marina Sova
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haim J. Wolfson
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Maayan Gal
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Ancuceanu R, Hovaneț MV, Cojocaru-Toma M, Anghel AI, Dinu M. Potential Antifungal Targets for Aspergillus sp. from the Calcineurin and Heat Shock Protein Pathways. Int J Mol Sci 2022; 23:ijms232012543. [PMID: 36293395 PMCID: PMC9603945 DOI: 10.3390/ijms232012543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Aspergillus species, especially A. fumigatus, and to a lesser extent others (A. flavus, A. niger, A. terreus), although rarely pathogenic to healthy humans, can be very aggressive to immunocompromised patients (they are opportunistic pathogens). Although survival rates for such infections have improved in recent decades following the introduction of azole derivatives, they remain a clinical challenge. The fact that current antifungals act as fungistatic rather than fungicide, that they have limited safety, and that resistance is becoming increasingly common make the need for new, more effective, and safer therapies to become more acute. Over the last decades, knowledge about the molecular biology of A. fumigatus and other Aspergillus species, and particularly of calcineurin, Hsp90, and their signaling pathway proteins, has progressed remarkably. Although calcineurin has attracted much interest, its adverse effects, particularly its immunosuppressive effects, make it less attractive than it might at first appear. The situation is not very different for Hsp90. Other proteins from their signaling pathways, such as protein kinases phosphorylating the four SPRR serine residues, CrzA, rcnA, pmcA-pmcC (particularly pmcC), rfeF, BAR adapter protein(s), the phkB histidine kinase, sskB MAP kinase kinase, zfpA, htfA, ctfA, SwoH (nucleoside diphosphate kinase), CchA, MidA, FKBP12, the K27 lysine position from Hsp90, PkcA, MpkA, RlmA, brlA, abaA, wetA, other heat shock proteins (Hsp70, Hsp40, Hsp12) currently appear promising and deserve further investigation as potential targets for antifungal drug development.
Collapse
Affiliation(s)
- Robert Ancuceanu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
- Correspondence: (R.A.); (M.V.H.)
| | - Marilena Viorica Hovaneț
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
- Correspondence: (R.A.); (M.V.H.)
| | - Maria Cojocaru-Toma
- Faculty of Pharmacy, Nicolae Testemițanu State University of Medicine and Pharmacy, 2025 Chisinau, Moldova
| | - Adriana-Iuliana Anghel
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Mihaela Dinu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
12
|
Jedlička M, Feglarová T, Janstová L, Hortová-Kohoutková M, Frič J. Lactate from the tumor microenvironment - A key obstacle in NK cell-based immunotherapies. Front Immunol 2022; 13:932055. [PMID: 36330529 PMCID: PMC9623302 DOI: 10.3389/fimmu.2022.932055] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/30/2022] [Indexed: 05/03/2025] Open
Abstract
Recent findings about the new roles of lactate have changed our understanding of this end product of glycolysis or fermentation that was once considered only a waste product. It is now well accepted that lactate acts as a signaling molecule and fuel source for cancer cells in a glucose-restricted environment. Moreover, lactate and lactate dehydrogenase are markers of poor prognosis of many cancers and regulate many functions of immune cells. The presence of lactate in the tumor microenvironment (TME) leads to polarization of the immunosuppressive phenotypes of dendritic cells and impairs the cytotoxic abilities of T cells and NK cells, and as such lactate is a major obstacle to immune-cell effector functions and the efficacy of cell-based immunotherapies. Emerging evidence suggests that lactate in the TME might be a novel therapeutic target to enhance the immunotherapeutic potential of cell-based therapies. This review describes our current understanding of the role of lactate in tumor biology, including its detrimental effects on cell-based immunotherapy in cancer. We also highlight how the role of lactate in the TME must be considered when producing cell therapies designed for adoptive transfer and describe how targeted modulation of lactate in the TME might boost immune-cell functions and positively impact cellular immunotherapy, with a focus on NK cell.
Collapse
Affiliation(s)
- Marek Jedlička
- Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
- Faculty of Science, Charles University in Prague, Prague, Czechia
| | - Tereza Feglarová
- Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Lucie Janstová
- Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
- Faculty of Science, Charles University in Prague, Prague, Czechia
| | - Marcela Hortová-Kohoutková
- Cellular and Molecular Immunoregulation Group, International Clinical Research Center of St. Anne´s University Hospital Brno, Brno, Czechia
| | - Jan Frič
- Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
- Cellular and Molecular Immunoregulation Group, International Clinical Research Center of St. Anne´s University Hospital Brno, Brno, Czechia
| |
Collapse
|
13
|
Copur S, Demiray A, Kanbay M. Uric acid in metabolic syndrome: Does uric acid have a definitive role? Eur J Intern Med 2022; 103:4-12. [PMID: 35508444 DOI: 10.1016/j.ejim.2022.04.022] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
Abstract
Increased serum uric acid (SUA) levels are commonly seen in patients with metabolic syndrome and are widely accepted as risk factors for hypertension, gout, non-alcoholic fatty liver disease, chronic kidney disease (CKD), and cardiovascular diseases. Although some ambiguity for the exact role of uric acid (UA) in these diseases is still present, several pathophysiological mechanisms have been identified such as increased oxidative stress, inflammation, and apoptosis. Accumulating evidence in genomics enlightens genetic variabilities and some epigenetic changes that can contribute to hyperuricemia. Here we discuss the role of UA within metabolism and the consequences of asymptomatic hyperuricemia while providing newfound evidence for the associations between UA and gut microbiota and vitamin D. Increased SUA levels and beneficial effects of lowering SUA levels need to be elucidated more to understand its complicated function within different metabolic pathways and set optimal target levels for SUA for reducing risks for metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
14
|
Colombo M, Marongiu L, Mingozzi F, Marzi R, Cigni C, Facchini FA, Rotem R, Valache M, Stucchi G, Rocca G, Gornati L, Rizzuto MA, Salvioni L, Zanoni I, Gori A, Prosperi D, Granucci F. Specific immunosuppressive role of nanodrugs targeting calcineurin in innate myeloid cells. iScience 2022; 25:105042. [PMID: 36124235 PMCID: PMC9482116 DOI: 10.1016/j.isci.2022.105042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/04/2022] [Accepted: 08/25/2022] [Indexed: 11/27/2022] Open
Abstract
Calcineurin (CN) inhibitors currently used to avoid transplant rejection block the activation of adaptive immune responses but also prevent the development of tolerance toward the graft, by directly inhibiting T cells. CN, through the transcription factors of the NFAT family, plays an important role also in the differentiation dendritic cells (DCs), the main cells responsible for the activation of T lymphocytes. Therefore, we hypothesized that the inhibition of CN only in DCs and not in T cells could be sufficient to prevent T cell responses, while allowing for the development of tolerance. Here, we show that inhibition of CN/NFAT pathway in innate myeloid cells, using a new nanoconjugate capable of selectively targeting phagocytes in vivo, protects against graft rejection and induces a longer graft acceptance compared to common CN inhibitors. We propose a new generation of nanoparticles-based selective immune suppressive agents for a better control of transplant acceptance. Calcineurin/NFATc2 pathway is required to enable DC migration to draining lymph nodes Calcineurin/NFATc2 pathway in DCs is required for type I immune responses activation Superparamagnetic iron oxide NPs can be used to efficiently target phagocytes in vivo Specific delivery of calcineurin inhibitor by NPs to phagocytes induce graft acceptance
Collapse
Affiliation(s)
- Miriam Colombo
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Laura Marongiu
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Francesca Mingozzi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Roberta Marzi
- Humabs BioMed, Bellinzona, Canton Ticino, Switzerland
| | - Clara Cigni
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Fabio Alessandro Facchini
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Rany Rotem
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Mihai Valache
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Giulia Stucchi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Giuseppe Rocca
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Laura Gornati
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Maria Antonietta Rizzuto
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Lucia Salvioni
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Division of Gastroenterology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alessandro Gori
- Istituto di Scienze e Tecnologie Chimiche, National Research Council of Italy (SCITEC-CNR), Via Mario Bianco, 9, 20131 Milan, Italy
| | - Davide Prosperi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
- Corresponding author
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
- Corresponding author
| |
Collapse
|
15
|
Chen L, Song M, Yao C. Calcineurin in development and disease. Genes Dis 2022; 9:915-927. [PMID: 35685477 PMCID: PMC9170610 DOI: 10.1016/j.gendis.2021.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Calcineurin (CaN) is a unique calcium (Ca2+) and calmodulin (CaM)-dependent serine/threonine phosphatase that becomes activated in the presence of increased intracellular Ca2+ level. CaN then functions to dephosphorylate target substrates including various transcription factors, receptors, and channels. Once activated, the CaN signaling pathway participates in the development of multiple organs as well as the onset and progression of various diseases via regulation of different cellular processes. Here, we review current literature regarding the structural and functional properties of CaN, highlighting its crucial role in the development and pathogenesis of immune system disorders, neurodegenerative diseases, kidney disease, cardiomyopathy and cancer.
Collapse
Affiliation(s)
- Lei Chen
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Min Song
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Chunyan Yao
- Department of Blood Transfusion, First Affiliated Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| |
Collapse
|
16
|
Li S, Zhou H, Xie M, Zhang Z, Gou J, Yang J, Tian C, Ma K, Wang CY, Lu Y, Li Q, Peng W, Xiang M. Regenerating islet-derived protein 3 gamma (Reg3g) ameliorates tacrolimus-induced pancreatic β-cell dysfunction in mice by restoring mitochondrial function. Br J Pharmacol 2022; 179:3078-3095. [PMID: 35060126 DOI: 10.1111/bph.15803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Tacrolimus a first-line medication used after transplantation can induce β-cell dysfunction, causing new-onset diabetes mellitus (NODM). Regenerating islet-derived protein 3 gamma (Reg3g), a member of the pancreatic regenerative gene family, has been reported to improve type 1 diabetes by promoting β-cell regeneration. We aim to investigate the role of Reg3g in reversing tacrolimus-induced β-cell dysfunction and NODM in mice. EXPERIMENTAL APPROACH Circulating REG3A (the human homologue of mouse Reg3g) in heart transplantation patients treated with tacrolimus was detected. The glucose-stimulated insulin secretion and mitochondrial functions, including mitochondria membrane potential (MMP), mitochondria calcium, ATP production, oxygen consumption rate and mitochondrial morphology were investigated in β-cells. Additionally, effects of Reg3g on tacrolimus-induced NODM in mice were analysed. KEY RESULTS Circulating REG3A level in heart transplantation patients with NODM significantly decreased compared with those without diabetes. Tacrolimus down-regulated Reg3g via inhibiting STAT3-mediated transcription activation. Moreover, Reg3g restored glucose-stimulated insulin secretion suppressed by tacrolimus in β-cells by improving mitochondrial functions, including increased MMP, mitochondria calcium uptake, ATP production, oxygen consumption rate and contributing to an intact mitochondrial morphology. Mechanistically, Reg3g increased accumulation of pSTAT3(Ser727) in mitochondria by activating ERK1/2-STAT3 signalling pathway, leading to restoration of tacrolimus-induced mitochondrial impairment. Reg3g overexpression also effectively mitigated tacrolimus-induced NODM in mice. CONCLUSION AND IMPLICATIONS Reg3g can significantly ameliorate tacrolimus-induced β-cell dysfunction by restoring mitochondrial function in a pSTAT3(Ser727)-dependent manner. Our observations identify a novel Reg3g-mediated mechanism that is involved in tacrolimus-induced NODM and establish the novel role of Reg3g in reversing tacrolimus-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyuan Xie
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zijun Zhang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Gou
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Ma
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yi Lu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Peng
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Gao R, Zhang Y, Zeng C, Li Y. The role of NFAT in the pathogenesis and targeted therapy of hematological malignancies. Eur J Pharmacol 2022; 921:174889. [DOI: 10.1016/j.ejphar.2022.174889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023]
|
18
|
Cross-tissue transcriptome-wide association studies identify susceptibility genes shared between schizophrenia and inflammatory bowel disease. Commun Biol 2022; 5:80. [PMID: 35058554 PMCID: PMC8776955 DOI: 10.1038/s42003-022-03031-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic correlations and an increased incidence of psychiatric disorders in inflammatory-bowel disease have been reported, but shared molecular mechanisms are unknown. We performed cross-tissue and multiple-gene conditioned transcriptome-wide association studies for 23 tissues of the gut-brain-axis using genome-wide association studies data sets (total 180,592 patients) for Crohn’s disease, ulcerative colitis, primary sclerosing cholangitis, schizophrenia, bipolar disorder, major depressive disorder and attention-deficit/hyperactivity disorder. We identified NR5A2, SATB2, and PPP3CA (encoding a target for calcineurin inhibitors in refractory ulcerative colitis) as shared susceptibility genes with transcriptome-wide significance both for Crohn’s disease, ulcerative colitis and schizophrenia, largely explaining fine-mapped association signals at nearby genome-wide association study susceptibility loci. Analysis of bulk and single-cell RNA-sequencing data showed that PPP3CA expression was strongest in neurons and in enteroendocrine and Paneth-like cells of the ileum, colon, and rectum, indicating a possible link to the gut-brain-axis. PPP3CA together with three further suggestive loci can be linked to calcineurin-related signaling pathways such as NFAT activation or Wnt. Florian Uellendahl-Werth et al. conduct cross-tissue transcriptome-wide association studies to explore genetic mechanisms shared across immune-related and psychiatric traits. Their results identify several genes (including PPP3CA) that could mediate the interplay between psychiatric and inflammatory disease.
Collapse
|
19
|
Ravichandran G, Sarkar P, Chen TW, Almaary KS, Elshikh MS, Elumalai P, Ramasamy H, Karuppiah K, Arockiaraj J. Antibacterial Effect of a Short Peptide, VV18, from Calcineurin-A of Macrobrachium rosenbergii: Antibiofilm Agent Against Escherichia coli and a Bacterial Membrane Disruptor in Pseudomonas aeruginosa. Int J Pept Res Ther 2022; 28:22. [DOI: 10.1007/s10989-021-10332-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 11/26/2022]
|
20
|
Spironolactone Inhibits Cardiomyocyte Hypertrophy by Regulating the Ca 2+/Calcineurin/p-NFATc3 Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3843830. [PMID: 34956570 PMCID: PMC8702305 DOI: 10.1155/2021/3843830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 01/19/2023]
Abstract
This study aimed to investigate the protective effect and molecular mechanism of spironolactone in isoproterenol-induced cardiomyocyte hypertrophy. In this study, primary cardiomyocytes were extracted from the heart of neonatal rats. After stable culture, they were processed with isoproterenol alone or isoproterenol (10 μM) combined with different doses (low dose of 10 μM and high dose of 50 μM), and the cellular activity was determined by MTT experiment. The volume of cells was measured with an inverted microscope and CIAS-1000 cell image analysis system. The mRNA expression levels of ANP and BNP in cells were explored by RT-qPCR. The levels of ANP and BNP proteins and NFATc3 phosphorylation in the nucleus were detected by western blot. The extracellular Ca2+ concentration and CaN activity were measured by colorimetry with the kit. Isoproterenol significantly enlarged the volume of cardiomyocytes (p < 0.001), upregulated mRNA and expression levels of ANP and BNP proteins (p < 0.001), increased extracellular Ca2+ concentration and CaN activity (p < 0.001), and upregulated NFATc3 phosphorylation in the nucleus (p < 0.001). The volume of cells treated with isoproterenol combined with different doses of spironolactone significantly decreased compared with those treated with isoproterenol alone (p < 0.001). mRNA and expression levels of ANP and BNP proteins downregulated significantly (p < 0.001). The extracellular Ca2+ (p < 0.01) concentration and CaN activity (p < 0.001) decreased significantly, and NFATc3 phosphorylation in the nucleus downregulated significantly (p < 0.001). There was no significant difference in cell volume (p=0.999), ANP and BNP mRNA (p=0.695), expression levels of proteins, CaN activity (0.154), and NFATc3 phosphorylation in the nucleus between the cells treated with isoproterenol combined with high-dose spironolactone and those in the control group. In conclusion, spironolactone can reverse isoproterenol-induced cardiomyocyte hypertrophy by inhibiting the Ca2+/CaN/NFATc3 pathway.
Collapse
|
21
|
Niu J, Qin B, Wang C, Chen C, Yang J, Shao H. Identification of Key Immune-Related Genes in the Progression of Septic Shock. Front Genet 2021; 12:668527. [PMID: 34804111 PMCID: PMC8595268 DOI: 10.3389/fgene.2021.668527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Septic shock is the severe complication of sepsis, with a high mortality. The inflammatory response regulates the immune status and mediates the progression of septic shock. In this study, we aim to identify the key immune-related genes (IRGs) of septic shock and explore their potential mechanism. Methods: Gene expression profiles of septic shock blood samples and normal whole blood samples were retrieved from the Gene Expression Omnibus (GEO) and Genotype-Tissue Expression Portal (GTEx). The differential expression genes (DEGs) and septic shock-specific immune-related genes (SSSIRGs) were evaluated and identified, along with the immune components by "cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT, version x)" algorithm. Additionally, in order to explore the key regulatory network, the relationship among SSSIRGs, upstream transcription factors (TFs), and downstream signaling pathways were also identified by Gene Set Variation Analysis (GSVA) and co-expression analysis. Moreover, the Connectivity Map (CMap) analysis was applied to find bioactive small molecules against the members of regulation network while Chromatin Immunoprecipitation sequencing (ChIP-seq) and Assay for Targeting Accessible-Chromatin with high-throughput sequencing (ATAC-seq) data were used to validate the regulation mechanism of the network. Results: A total of 14,843 DEGs were found between 63 septic shock blood samples and 337 normal whole blood samples. Then, we identified septic shock-specific 839 IRGs as the intersection of DEGs and IRGs. Moreover, we uncovered the regulatory networks based on co-expression analysis and found 28 co-expression interaction pairs. In the regulation network, protein phosphatase 3, catalytic subunit, alpha isozyme (PPP3CA) may regulate late estrogen response, glycolysis and TNFα signaling via NFκB and HLA; Kirsten rat sarcoma viral oncogene homolog (KRAS) may be related to late estrogen response and HLA; and Toll-like receptor 8 (TLR8) may be associated with TNFα signaling via NFκB. And the regulation mechanisms between TFs and IRGs (TLR8, PPP3CA, and KRAS) were validated by ChIP-seq and ATAC-seq. Conclusion: Our data identify three SSSIRGs (TLR8, PPP3CA, and KRAS) as candidate therapeutic targets for septic shock and provide constructed regulatory networks in septic shock to explore its potential mechanism.
Collapse
Affiliation(s)
- Jingjing Niu
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Bingyu Qin
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Cunzhen Wang
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Chao Chen
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Jianxu Yang
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Huanzhang Shao
- Department of Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| |
Collapse
|
22
|
Vymazal O, Bendíčková K, De Zuani M, Vlková M, Hortová-Kohoutková M, Frič J. Immunosuppression Affects Neutrophil Functions: Does Calcineurin-NFAT Signaling Matter? Front Immunol 2021; 12:770515. [PMID: 34795676 PMCID: PMC8593005 DOI: 10.3389/fimmu.2021.770515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Neutrophils are innate immune cells with important roles in antimicrobial defense. However, impaired or dysregulated neutrophil function can result in host tissue damage, loss of homeostasis, hyperinflammation or pathological immunosuppression. A central link between neutrophil activation and immune outcomes is emerging to be the calcineurin-nuclear factor of activated T cells (NFAT) signaling pathway, which is activated by neutrophil detection of a microbial threat via pattern recognition receptors and results in inflammatory cytokine production. This potent pro-inflammatory pathway is also the target of several immunosuppressive drugs used for the treatment of autoimmune disorders, during solid organ and hematopoietic cell transplantations, and as a part of anti-cancer therapy: but what effects these drugs have on neutrophil function, and their broader consequences for immune homeostasis and microbial defense are not yet known. Here, we bring together the emerging literature describing pathology- and drug- induced neutrophil impairment, with particular focus on their effects on calcineurin-NFAT signaling in the innate immune compartment.
Collapse
Affiliation(s)
- Ondřej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Marco De Zuani
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Marcela Vlková
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Clinical Immunology and Allergology, St. Anne´s University Hospital, Brno, Czechia
| | | | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia.,Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
| |
Collapse
|
23
|
Abstract
Acute myeloid leukaemia (AML) is a haematological cancer with poor outcomes due to a lack of efficacious targeted therapies. The Nuclear Factor of Activated T Cells (NFAT) family of transcription factors is well characterised as a regulator of the cell cycle and differentiation in the myeloid lineage. Recent evidence has demonstrated that NFAT family members may have roles in regulating AML leukemogenesis and resistance to targeted therapy in myeloid leukaemia. Furthermore, gene expression data from patient samples show that some NFATs are more highly expressed in poorly differentiated AML and after disease relapse, implying that the NFAT family may have roles in specific types of AML. This review outlines the evidence for the role of NFAT in healthy myeloid tissue and explores how NFAT might regulate AML pathogenesis, highlighting the potential to target specific NFAT proteins therapeutically in AML.
Collapse
|
24
|
Wang S, Qian H, Zhang L, Liu P, Zhuang D, Zhang Q, Bai F, Wang Z, Yan Y, Guo J, Huang J, Wu X. Inhibition of Calcineurin/NFAT Signaling Blocks Oncogenic H-Ras Induced Autophagy in Primary Human Keratinocytes. Front Cell Dev Biol 2021; 9:720111. [PMID: 34350189 PMCID: PMC8328491 DOI: 10.3389/fcell.2021.720111] [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: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Mutations of H-Ras, a member of the RAS family, are preferentially found in cutaneous squamous cell carcinomas (SCCs). H-Ras has been reported to induce autophagy, which plays an essential role in tissue homeostasis in multiple types of cancer cells and in fibroblasts, however, the potential role of H-Ras in regulating autophagy in human keratinocytes has not been reported. In this study, we found that the stable expression of the G12V mutant of H-RAS (H-Ras G12V ) induced autophagy in human keratinocytes, and interestingly, the induction of autophagy was strongly blocked by inhibiting the calcineurin/nuclear factor of activated T cells (NFAT) pathway with either a calcineurin inhibitor (Cyclosporin A) or a NFAT inhibitor (VIVIT), or by the small interfering RNA (siRNA) mediated knockdown of calcineurin B1 or NFATc1 in vitro, as well as in vivo. To characterize the role of the calcineurin/NFAT pathway in H-Ras induced autophagy, we found that H-Ras G12V promoted the nuclear translocation of NFATc1, an indication of the activation of the calcineurin/NFAT pathway, in human keratinocytes. However, activation of NFATc1 either by the forced expression of NFATc1 or by treatment with phenformin, an AMPK activator, did not increase the formation of autophagy in human keratinocytes. Further study revealed that inhibiting the calcineurin/NFAT pathway actually suppressed H-Ras expression in H-Ras G12V overexpressing cells. Finally, chromatin immunoprecipitation (ChIP) assays showed that NFATc1 potentially binds the promoter region of H-Ras and the binding efficiency was significantly enhanced by the overexpression of H-Ras G12V , which was abolished by treatment with the calcineurin/NFAT pathway inhibitors cyclosporine A (CsA) or VIVIT. Taking these data together, the present study demonstrates that the calcineurin/NFAT signaling pathway controls H-Ras expression and interacts with the H-Ras pathway, involving the regulation of H-Ras induced autophagy in human keratinocytes.
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hua Qian
- Department of Stomatology, The Second Hospital of Shandong University, Jinan, China
| | - Liwei Zhang
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Panpan Liu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dexuan Zhuang
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Qun Zhang
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Fuxiang Bai
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zhihong Wang
- Qilu Children's Hospital of Shandong University, Jinan, China
| | - Yonggan Yan
- Center for Advanced Jet Engineering Technologies, Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, China
| | - Jing Guo
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Savaid Stomatology School of Hangzhou Medical College, Ningbo Stomatology Hospital, Ningbo, China
| | - Jun Huang
- Center for Advanced Jet Engineering Technologies, Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, China
| | - Xunwei Wu
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Banki E, Fisi V, Moser S, Wengi A, Carrel M, Loffing-Cueni D, Penton D, Kratschmar DV, Rizzo L, Lienkamp S, Odermatt A, Rinschen MM, Loffing J. Specific disruption of calcineurin-signaling in the distal convoluted tubule impacts the transcriptome and proteome, and causes hypomagnesemia and metabolic acidosis. Kidney Int 2021; 100:850-869. [PMID: 34252449 DOI: 10.1016/j.kint.2021.06.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/27/2022]
Abstract
Adverse effects of calcineurin inhibitors (CNI), such as hypertension, hyperkalemia, acidosis, hypomagnesemia and hypercalciuria, have been linked to dysfunction of the distal convoluted tubule (DCT). To test this, we generated a mouse model with an inducible DCT-specific deletion of the calcineurin regulatory subunit B alpha (CnB1-KO). Three weeks after CnB1 deletion, these mice exhibited hypomagnesemia and acidosis, but no hypertension, hyperkalemia or hypercalciuria. Consistent with the hypomagnesemia, CnB1-KO mice showed a downregulation of proteins implicated in DCT magnesium transport, including TRPM6, CNNM2, SLC41A3 and parvalbumin but expression of calcium channel TRPV5 in the kidney was unchanged. The abundance of the chloride/bicarbonate exchanger pendrin was increased, likely explaining the acidosis. Plasma aldosterone levels, kidney renin expression, abundance of phosphorylated sodium chloride-cotransporter and abundance of the epithelial sodium channel were similar in control and CnB1-KO mice, consistent with a normal sodium balance. Long-term potassium homeostasis was maintained in CnB1-KO mice, but in-vivo and ex-vivo experiments indicated that CnB1 contributes to acute regulation of potassium balance and sodium chloride-cotransporter. Tacrolimus treatment of control and CnB1-KO mice demonstrated that CNI-related hypomagnesemia is linked to impaired calcineurin-signaling in DCT, while hypocalciuria and hyponatremia occur independently of CnB1 in DCT. Transcriptome and proteome analyses of isolated DCTs demonstrated that CnB1 deletion impacts the expression of several DCT-specific proteins and signaling pathways. Thus, our data support a critical role of calcineurin for DCT function and provide novel insights into the pathophysiology of CNI side-effects and involved molecular players in the DCT.
Collapse
Affiliation(s)
- Eszter Banki
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Viktoria Fisi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sandra Moser
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Agnieszka Wengi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Monique Carrel
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - David Penton
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Denise V Kratschmar
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland
| | - Ludovica Rizzo
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Soeren Lienkamp
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland
| | - Markus M Rinschen
- Kidney Research Center, University of Cologne, Köln, Germany; Department of Biomedicine, Aarhus University, Aarhus, Denmark; III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Swiss National Centre for Competence in Research "Kidney Control of Homeostasis," Zurich, Switzerland.
| |
Collapse
|
26
|
Zhang X, Li G, Deng Q, Xu Z, Cen J, Xu J. Vomifoliol isolated from mangrove plant Ceriops tagal inhibits the NFAT signaling pathway with CN as the target enzyme in vitro. Bioorg Med Chem Lett 2021; 48:128235. [PMID: 34216746 DOI: 10.1016/j.bmcl.2021.128235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 06/17/2021] [Accepted: 06/26/2021] [Indexed: 11/26/2022]
Abstract
Vomifoliol, a natural sesquiterpene compound, is a secondary metabolite isolated from the mangrove plant Ceriops tagal. The present study aimed to determine the immunosuppressive effects and underlying mechanisms of vomifoliol on Jurkat cells in vitro. The results show that vomifoliol significantly inhibited calcineurin (CN) at concentrations resulting in relatively low cytotoxicity. Moreover, vomifoliol was found to exert an inhibitory effect on phorbol 12-myristate 13-acetate (PMA)/ ionomycin (Io) -induced Jurkat cells and the dephosphorylation of NFAT1. In addition, it reduced the expression of IL-2. Based on these results, we concluded that vomifoliol may inhibit the immune response of Jurkat cells, and vomifoliol may use CN as the target enzyme to inhibit NFAT signaling pathway. Therefore, vomifoliol may be promising as a low-toxic natural immunosuppressant.
Collapse
Affiliation(s)
- Xuexia Zhang
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China
| | - Gang Li
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China
| | - Qin Deng
- College of Horticulture and Landscape, Hainan University, Haikou 570228, PR China
| | - Zhiyong Xu
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, PR China
| | - Juren Cen
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, PR China.
| | - Jing Xu
- Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, School of Chemical Engineering and Technology, Hainan University, Haikou 570228, PR China.
| |
Collapse
|
27
|
Cellular Calcium Levels Influenced by NCA-2 Impact Circadian Period Determination in Neurospora. mBio 2021; 12:e0149321. [PMID: 34182778 PMCID: PMC8262947 DOI: 10.1128/mbio.01493-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intracellular calcium signaling has been implicated in the control of a variety of circadian processes in animals and plants, but its role in microbial clocks has remained largely cryptic. To examine the role of intracellular Ca2+ in the Neurospora clock, we screened mutants with knockouts of calcium transporter genes and identified a gene encoding a calcium exporter, nca-2, uniquely as having significant period effects. The loss of NCA-2 results in an increase in the cytosolic calcium level, and this leads to hyper-phosphorylation of core clock components, FRQ and WC-1, and a short period, as measured by both the core oscillator and the overt clock. Genetic analyses showed that mutations in certain frq phospho-sites and in Ca2+-calmodulin-dependent kinase 2 (camk-2) are epistatic to nca-2 in controlling the pace of the oscillator. These data are consistent with a model in which elevated intracellular Ca2+ leads to the increased activity of CAMK-2, leading to enhanced FRQ phosphorylation, accelerated closure of the circadian feedback loop, and a shortened circadian period length. At a mechanistic level, some CAMKs undergo more auto-phosphorylations in the Δnca-2 mutant, consistent with high calcium levels in the Δnca-2 mutant influencing the enzymatic activities of CAMKs. NCA-2 interacts with multiple proteins, including CSP-6, a protein known to be required for circadian output. Most importantly, the expression of nca-2 is circadian clock-controlled at both the transcriptional and translational levels, and this in combination with the period effects seen in strains lacking NCA-2 firmly places calcium signaling within the larger circadian system, where it acts as both an input to and an output from the core clock.
Collapse
|
28
|
NFAT signaling in human mesenchymal stromal cells affects extracellular matrix remodeling and antifungal immune responses. iScience 2021; 24:102683. [PMID: 34195564 PMCID: PMC8233198 DOI: 10.1016/j.isci.2021.102683] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/18/2021] [Accepted: 05/31/2021] [Indexed: 01/15/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) combined with calcineurin-nuclear factor of activated T cell (CN-NFAT) inhibitors are being tested as a treatment for graft-versus-host disease (GvHD). The immunosuppressive properties of MSCs seem beneficial; however, their response during fungal infection, which is an important cause of mortality in patients with GvHD , is unknown. We report that MSCs phagocytose the fungal component zymosan, resulting in phosphorylation of spleen tyrosine kinase (Syk), increase in cytosolic calcium levels, and ultimately, increase in NFAT1 nuclear translocation. RNA sequencing analysis of zymosan-treated MSCs showed that CN-NFAT inhibition affects extracellular matrix (ECM) genes but not cytokine expression that is under the control of the NF-κB pathway. When coculturing MSCs or decellularized MSC-ECM with human peripheral blood mononuclear cells (PBMCs), selective NFAT inhibition in MSCs decreased cytokine expression by PBMCs. These findings reveal a dual mechanism underlying the MSC response to zymosan: while NF-κB directly controls inflammatory cytokine expression, NFAT impacts immune-cell functions by regulating ECM remodeling. Stimulation of MSCs with zymosan activates NFAT and NF-kB via the dectin1-Syk axis Calcineurin-NFAT inhibition impacts the expression of extracellular matrix genes NF-kB pathway regulates cytokine expression in zymosan-stimulated MSCs Selective NFAT inhibition in MSCs impacts cytokine secretion of MSC-PBMC cocultures
Collapse
|
29
|
Fakhri S, Nouri Z, Moradi SZ, Akkol EK, Piri S, Sobarzo-Sánchez E, Farzaei MH, Echeverría J. Targeting Multiple Signal Transduction Pathways of SARS-CoV-2: Approaches to COVID-19 Therapeutic Candidates. Molecules 2021; 26:2917. [PMID: 34068970 PMCID: PMC8156180 DOI: 10.3390/molecules26102917] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Due to the complicated pathogenic pathways of coronavirus disease 2019 (COVID-19), related medicinal therapies have remained a clinical challenge. COVID-19 highlights the urgent need to develop mechanistic pathogenic pathways and effective agents for preventing/treating future epidemics. As a result, the destructive pathways of COVID-19 are in the line with clinical symptoms induced by severe acute coronary syndrome (SARS), including lung failure and pneumonia. Accordingly, revealing the exact signaling pathways, including inflammation, oxidative stress, apoptosis, and autophagy, as well as relative representative mediators such as tumor necrosis factor-α (TNF-α), nuclear factor erythroid 2-related factor 2 (Nrf2), Bax/caspases, and Beclin/LC3, respectively, will pave the road for combating COVID-19. Prevailing host factors and multiple steps of SARS-CoV-2 attachment/entry, replication, and assembly/release would be hopeful strategies against COVID-19. This is a comprehensive review of the destructive signaling pathways and host-pathogen interaction of SARS-CoV-2, as well as related therapeutic targets and treatment strategies, including potential natural products-based candidates.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
| | - Zeinab Nouri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, Ankara 06330, Turkey;
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; (S.F.); (S.Z.M.); (S.P.)
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9170022, Chile
| |
Collapse
|
30
|
Marin W, Marin D, Ao X, Liu Y. Mitochondria as a therapeutic target for cardiac ischemia‑reperfusion injury (Review). Int J Mol Med 2020; 47:485-499. [PMID: 33416090 PMCID: PMC7797474 DOI: 10.3892/ijmm.2020.4823] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myocardial infarction is the leading cause of cardiovascular-related mortality and chronic heart failure worldwide. As regards treatment, the reperfusion of ischemic tissue generates irreversible damage to the myocardium, which is termed 'cardiac ischemia-reperfusion (IR) injury'. Due to the large number of mitochondria in cardiomyocytes, an increasing number of studies have focused on the roles of mitochondria in IR injury. The primary causes of IR injury are reduced oxidative phosphorylation during hypoxia and the increased production of reactive oxygen species (ROS), together with the insufficient elimination of these oxidative species following reperfusion. IR injury includes the oxidation of DNA, incorrect modifications of proteins, the disruption of the mitochondrial membrane and respiratory chain, the loss of mitochondrial membrane potential (∆Ψm), Ca2+ over-load, mitochondrial permeability transition pore formation, swelling of the mitochondria, and ultimately, cardiomyocyte necrosis. The present review article discusses the molecular mechanisms of IR injury, and summarizes the metabolic and dynamic changes occurring in the mitochondria in response to IR stress. The mitochondria are strongly recommended as a target for the development of therapeutic agents; however, the appropriate use of agents remains a challenge.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Dennis Marin
- Qingdao University of Science and Technology, Qingdao, Shandong 266061, P.R. China
| | - Xiang Ao
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
31
|
Martinez-Chantar ML, Foti M. NFATc4: New hub in NASH development. J Hepatol 2020; 73:1313-1315. [PMID: 33032848 DOI: 10.1016/j.jhep.2020.08.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/14/2023]
Affiliation(s)
- Maria L Martinez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland; Diabetes Centre, Faculty of Medicine, University of Geneva, Switzerland; Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
32
|
Poulsen NN, von Brunn A, Hornum M, Blomberg Jensen M. Cyclosporine and COVID-19: Risk or favorable? Am J Transplant 2020; 20:2975-2982. [PMID: 32777170 PMCID: PMC7436557 DOI: 10.1111/ajt.16250] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/06/2020] [Accepted: 07/27/2020] [Indexed: 01/25/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is declared a global health emergency. COVID-19 is triggered by a novel coronavirus: severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Baseline characteristics of admitted patients with COVID-19 show that adiposity, diabetes, and hypertension are risk factors for developing severe disease, but so far immunosuppressed patients who are listed as high-risk patients have not been more susceptible to severe COVID-19 than the rest of the population. Multiple clinical trials are currently being conducted, which may identify more drugs that can lower mortality, morbidity, and burden on the society. Several independent studies have convincingly shown that cyclosporine inhibit replication of several different coronaviruses in vitro. The cyclosporine-analog alisporivir has recently been shown to inhibit SARS-CoV-2 in vitro. These findings are intriguing, although there is no clinical evidence for a protective effect to reduce the likelihood of severe COVID-19 or to treat the immune storm or acute respiratory distress syndrome (ARDS) that often causes severe morbidity. Here, we review the putative link between COVID-19 and cyclosporine, while we await more robust clinical data.
Collapse
Affiliation(s)
- Nadia Nicholine Poulsen
- Department of Growth and Reproduction, Group of Skeletal, Mineral, and Gonadal Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Albrecht von Brunn
- Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich/German Center for Infection Research (DZIF), Munich, Germany
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Department of Growth and Reproduction, Group of Skeletal, Mineral, and Gonadal Endocrinology, Rigshospitalet, Copenhagen, Denmark,Division of Bone and Mineral Research, HSDM/HMS Harvard University, Boston, MA, USA,Correspondence Martin Blomberg Jensen
| |
Collapse
|
33
|
Fedele AO, Carraro V, Xie J, Averous J, Proud CG. Cyclosporin A but not FK506 activates the integrated stress response in human cells. J Biol Chem 2020; 295:15134-15143. [PMID: 32843478 DOI: 10.1074/jbc.ra120.014531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/28/2020] [Indexed: 11/06/2022] Open
Abstract
Cyclosporin A (CsA) and tacrolimus (FK506) are valuable immunosuppressants for a range of clinical settings, including (but not limited to) organ transplantation and the treatment of autoimmune diseases. They function by inhibiting the activity of the Ca2+/calmodulin-dependent phosphatase calcineurin toward nuclear factor of activated T-cells (NF-AT) in T-lymphocytes. However, use of CsA is associated with more serious side effects and worse clinical outcomes than FK506. Here we show that CsA, but not FK506, causes activation of the integrated stress response (ISR), an event which is normally an acute reaction to various types of intracellular insults, such as nutrient deficiency or endoplasmic reticulum stress. These effects of CsA involve at least two of the stress-activated protein kinases (GCN2 and PERK) that act on the translational machinery to slow down protein synthesis via phosphorylation of the eukaryotic initiation factor (eIF) 2α and thereby induce the ISR. These actions of CsA likely contribute to the adverse effects associated with its clinical application.
Collapse
Affiliation(s)
- Anthony O Fedele
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health & Medical Research Institute, North Terrace Campus, Adelaide, Australia
| | - Valérie Carraro
- INRAE Unité de Nutrition Humaine, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Jianling Xie
- Lifelong Health Theme, South Australian Health & Medical Research Institute, North Terrace Campus, Adelaide, Australia
| | - Julien Averous
- INRAE Unité de Nutrition Humaine, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Christopher G Proud
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health & Medical Research Institute, North Terrace Campus, Adelaide, Australia; Lifelong Health Theme, South Australian Health & Medical Research Institute, North Terrace Campus, Adelaide, Australia; School of Biological Sciences, University of Adelaide, North Terrace Campus, Adelaide, Australia.
| |
Collapse
|
34
|
Li HF, Wu YL, Tseng TL, Chao SW, Lin H, Chen HH. Inhibition of miR-155 potentially protects against lipopolysaccharide-induced acute lung injury through the IRF2BP2-NFAT1 pathway. Am J Physiol Cell Physiol 2020; 319:C1070-C1081. [PMID: 33052070 DOI: 10.1152/ajpcell.00116.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sepsis-induced lung injury is a lethal complication with no effective treatment options, affecting millions of people worldwide. Oroxylin A (OroA) is a natural flavonoid with potent anticancer effects, but its modulating effect on inflammation through microRNAs (miRs) is not apparent. In this report, we investigated the target genes of the miR pathway mediated by OroA and assessed the potential for novel treatments of septic lung injury. An miR array screening and quantitative polymerase chain reaction identified that miR-155-5p could be a candidate regulated by OroA. Bioinformatics analysis indicated that interferon regulatory factor-2-binding protein-2 (IRF2BP2) might be a target of miR-155-5p, and this hypothesis was verified through reporter assays. In addition, an immunoprecipitation assay demonstrated that OroA increased the binding activity of IRF2BP2 to the nuclear factor of activated T-cells 1 (NFAT1), causing inducible nitric oxide synthase to cause an inflammatory reaction. Finally, the direct injection of short hairpin RNA (shRNA)-miR-155-5p into the bone marrow of mice ameliorated LPS-induced acute lung injury and inflammation in mice. Our results provide new mechanistic insights into the role of the OroA-induced miR-155-5p-IRF2BP2-NFAT1 axis in sepsis, demonstrating that direct bone marrow injection of lentivirus containing shRNA-155-5p could prove to be a potential future clinical application in alleviating sepsis-induced acute lung injury.
Collapse
Affiliation(s)
- Hsiao-Fen Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,PhD Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lin Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Tzu-Ling Tseng
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shih-Wei Chao
- Drug Development Center, China Medical University, Taichung, Taiwan
| | - Heng Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,PhD Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
35
|
Kumar V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol 2020; 89:107087. [PMID: 33075714 PMCID: PMC7550173 DOI: 10.1016/j.intimp.2020.107087] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022]
Abstract
Sepsis infects more than 48.9 million people world-wide, with 19.7 million deaths. Cytokine storm plays a significant role in sepsis, along with severe COVID-19. TLR signaling pathways plays a crucial role in generating the cytokine storm. Endogenous negative regulators of TLR signaling are crucial to regulate cytokine storm.
Cytokine storm generates during various systemic acute infections, including sepsis and current pandemic called COVID-19 (severe) causing devastating inflammatory conditions, which include multi-organ failure or multi-organ dysfunction syndrome (MODS) and death of the patient. Toll-like receptors (TLRs) are one of the major pattern recognition receptors (PRRs) expressed by immune cells as well as non-immune cells, including neurons, which play a crucial role in generating cytokine storm. They recognize microbial-associated molecular patterns (MAMPs, expressed by pathogens) and damage or death-associate molecular patterns (DAMPs; released and/expressed by damaged/killed host cells). Upon recognition of MAMPs and DAMPs, TLRs activate downstream signaling pathways releasing several pro-inflammatory mediators [cytokines, chemokines, interferons, and reactive oxygen and nitrogen species (ROS or RNS)], which cause acute inflammation meant to control the pathogen and repair the damage. Induction of an exaggerated response due to genetic makeup of the host and/or persistence of the pathogen due to its evasion mechanisms may lead to severe systemic inflammatory condition called sepsis in response to the generation of cytokine storm and organ dysfunction. The activation of TLR-induced inflammatory response is hardwired to the induction of several negative feedback mechanisms that come into play to conclude the response and maintain immune homeostasis. This state-of-the-art review describes the importance of TLR signaling in the onset of the sepsis-associated cytokine storm and discusses various host-derived endogenous negative regulators of TLR signaling pathways. The subject is very important as there is a vast array of genes and processes implicated in these negative feedback mechanisms. These molecules and mechanisms can be targeted for developing novel therapeutic drugs for cytokine storm-associated diseases, including sepsis, severe COVID-19, and other inflammatory diseases, where TLR-signaling plays a significant role.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
36
|
Shi Y, Shu H, Wang X, Zhao H, Lu C, Lu A, He X. Potential Advantages of Bioactive Compounds Extracted From Traditional Chinese Medicine to Inhibit Bone Destructions in Rheumatoid Arthritis. Front Pharmacol 2020; 11:561962. [PMID: 33117162 PMCID: PMC7577042 DOI: 10.3389/fphar.2020.561962] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Bone destruction is an important pathological feature of rheumatoid arthritis (RA), which finally leads to the serious decline of life quality in RA patients. Bone metabolism imbalance is the principal factor of bone destruction in RA, which is manifested by excessive osteoclast-mediated bone resorption and inadequate osteoblast-mediated bone formation. Although current drugs alleviate the process of bone destruction to a certain extent, there are still many deficiencies. Recent studies have shown that traditional Chinese medicine (TCM) could effectively suppress bone destruction of RA. Some bioactive compounds from TCM have shown good effect on inhibiting osteoclast differentiation and promoting osteoblast proliferation. This article reviews the research progress of bioactive compounds exacted from TCM in inhibiting bone destruction of RA, so as to provide references for further clinical and scientific research.
Collapse
Affiliation(s)
- Yingjie Shi
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyang Shu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyu Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Hanxiao Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Chinese Medicine, Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, Hong Kong Baptist University, Hong Kong, Hong Kong
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Roberts MB, Fishman JA. Immunosuppressive Agents and Infectious Risk in Transplantation: Managing the "Net State of Immunosuppression". Clin Infect Dis 2020; 73:e1302-e1317. [PMID: 32803228 DOI: 10.1093/cid/ciaa1189] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Successful solid organ transplantation reflects meticulous attention to the details of immunosuppression, balancing risks for graft rejection against risks for infection. The 'net state of immune suppression' is a conceptual framework of all factors contributing to infectious risk. Assays which measure immune function in the immunosuppressed transplant recipient relative to infectious risk and allograft function are lacking. The best measures of integrated immune function may be quantitative viral loads to assess the individual's ability to control latent viral infections. Few studies address adjustment of immunosuppression during active infections. Thus, confronted with infection in solid organ recipients, the management of immunosuppression is based largely on clinical experience. This review examines known measures of immune function and the immunologic effects of common immunosuppressive drugs and available studies reporting modification of drug regimens for specific infections. These data provide a conceptual framework for the management of immunosuppression during infection in organ recipients.
Collapse
Affiliation(s)
- Matthew B Roberts
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston MA
| | - Jay A Fishman
- Transplant Infectious Disease and Compromised Host Program and Transplant Center, Massachusetts General Hospital, Boston MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
38
|
Luo W, Luo Y, Yang J. Proteomics-based screening of the target proteins associated with antidepressant-like effect and mechanism of nimesulide. Sci Rep 2020; 10:11052. [PMID: 32632112 PMCID: PMC7338510 DOI: 10.1038/s41598-020-66420-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/19/2020] [Indexed: 11/25/2022] Open
Abstract
Nimesulide is an inhibitor of COX-2 with antioxidant and anti-inflammatory effects. However, few studies have explored the antidepressant mechanism of nimesulide. Here, we evaluated the therapeutic effects of nimesulide on CUMS rats. iTRAQ technology was used to identify the differentially expressed protein in the hippocampus between CUMS and nimesulide-treated rats to identify the possible molecular mechanism of its effects. We found that nimesulide had positive effects on depressive-like behaviors and inflammatory factors in depressed rats. Using proteomics technologies, we screened 16 differentially expressed proteins in CUMS-exposed rats after nimesulide treatment, 5 of which were related to inflammation. Overall, these results show that nimesulide might mediate its antidepressant effect on depressed rats through the inhibition of oxidative stress inflammatory response.
Collapse
Affiliation(s)
- Wen Luo
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China
| | - Ying Luo
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China
| | - Junqing Yang
- College of Pharmacy, Chongqing Medical University, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| |
Collapse
|
39
|
Zhang K, Wang W, Chen L, Liu Y, Hu J, Guo F, Tian W, Wang Y, Xue F. Cross‑validation of genes potentially associated with neoadjuvant chemotherapy and platinum‑based chemoresistance in epithelial ovarian carcinoma. Oncol Rep 2020; 44:909-926. [PMID: 32705213 PMCID: PMC7388274 DOI: 10.3892/or.2020.7668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
Ovarian carcinomas have the poorest prognosis and the highest mortality among gynecological malignancies. Neoadjuvant chemotherapy (NACT) is considered as a novel therapeutic strategy and an alternative treatment for advanced epithelial ovarian cancer (AEOC). The aim of the present study was to identify the core genes related to platinum‑based NACT resistance in AEOC and to allow screening at the molecular level for the most appropriate ovarian cancer patients for NACT. We obtained three drug‑resistant microarrays GSE114206, GSE41499 and GSE33482 from the Gene Expression Omnibus (GEO) database as well as a microarray representing NACT, GSE109934. Bioinformatics analysis revealed the nature of the four potential candidate genes for using in functional enrichment analyses and interaction network construction. The potential associations and possible genetic alterations among the DEGs were summarized using the STRING database in Cytoscape and the cBioPortal visualization tool, respectively. A total of 63 genes were identified as DEGs from GSE109934 representing NACT. From the drug‑resistant GSE114206 and GSE41499 datasets, 106 DEGs containing 36 upregulated genes and 70 downregulated genes were selected, and from the drug‑resistant GSE114206 and GSE33482 datasets, 406 DEGs with 157 upregulated genes and 249 downregulated genes were selected. The 36 upregulated DEGs and the 70 downregulated genes were notably abundant in the different categories. In KEGG pathway analysis, the 157 upregulated genes and the 249 downregulated genes were concentrated in distinctive signaling pathways. Four potential genes associated with NACT and platinum‑based chemoresistance were screened, including nuclear factor of activated T‑cells, cytoplasmic 1 (NAFTc1), Kruppel‑like factor 4 (KLF4), nuclear receptor subfamily 4 group A member 3 (NR4A3) and hepatocyte growth factor (HGF). Our study showed that the mRNA expression levels of NAFTc1, NR4A3 and HGF were increased in drug‑resistant OC cell lines (all P<0.01), whereas the mRNA expression levels of KLF4 were notably lower in the SKOV3‑CDDP and HeyA8‑CDDP cell line (all P<0.01) but higher in the A2780‑CBP cell line. The NAFTc1, KLF4, NR4A3 and HGF genes may be potential therapeutic targets for NACT and platinum‑based chemoresistance factors as well as candidate biomarkers in AEOC. Determination of the expression levels of these four genes in tumor tissues before planning NACT treatment or initial surgery would be beneficial for AEOC patients.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Weihan Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro‑Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Lingli Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yulin Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jiali Hu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Fei Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
40
|
Bendickova K, Fric J. Roles of IL-2 in bridging adaptive and innate immunity, and as a tool for cellular immunotherapy. J Leukoc Biol 2020; 108:427-437. [PMID: 32480431 PMCID: PMC7384134 DOI: 10.1002/jlb.5mir0420-055r] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/01/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
IL-2 was initially characterized as a T cell growth factor in the 1970s, and has been studied intensively ever since. Decades of research have revealed multiple and diverse roles for this potent cytokine, indicating a unique linking role between adaptive and innate arms of the immune system. Here, we review the literature showing that IL-2 is expressed in a plethora of cell types across the immune system, where it has indispensable functions in orchestrating cellular interactions and shaping the nature and magnitude of immune responses. Emerging from the basic research that has revealed the molecular mechanisms and the complexity of the biologic actions of IL-2, several immunotherapeutic approaches have now focused on manipulating the levels of this cytokine in patients. These strategies range from inhibition of IL-2 to achieve immunosuppression, to the application of IL-2 as a vaccine adjuvant and in cancer therapies. This review will systematically summarize the major findings in the field and identify key areas requiring further research in order to realize the potential of IL-2 in the treatment of human diseases.
Collapse
Affiliation(s)
- Kamila Bendickova
- International Clinical Research CenterSt. Anne's University Hospital BrnoBrnoCzech Republic
| | - Jan Fric
- International Clinical Research CenterSt. Anne's University Hospital BrnoBrnoCzech Republic
- Institute of Hematology and Blood TransfusionPragueCzech Republic
| |
Collapse
|
41
|
Wang Z, Zhao Y, Phipps-Green A, Liu-Bryan R, Ceponis A, Boyle DL, Wang J, Merriman TR, Wang W, Terkeltaub R. Differential DNA Methylation of Networked Signaling, Transcriptional, Innate and Adaptive Immunity, and Osteoclastogenesis Genes and Pathways in Gout. Arthritis Rheumatol 2020; 72:802-814. [PMID: 31738005 DOI: 10.1002/art.41173] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In gout, autoinflammatory responses to urate crystals promote acute arthritis flares, but the pathogeneses of tophi, chronic synovitis, and erosion are less well understood. Defining the pathways of epigenomic immunity training can reveal novel pathogenetic factors and biomarkers. The present study was undertaken to seminally probe differential DNA methylation patterns utilizing epigenome-wide analyses in patients with gout. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from a San Diego cohort of patients with gout (n = 16) and individually matched healthy controls (n = 14). PBMC methylome data were processed with ChAMP package in R. ENCODE data and Taiji data analysis software were used to analyze transcription factor (TF)-gene networks. As an independent validation cohort, whole blood DNA samples from New Zealand Māori subjects (n = 13 patients with gout, n = 16 control subjects without gout) were analyzed. RESULTS Differentially methylated loci clearly separated gout patients from controls, as determined by hierarchical clustering and principal components analyses. IL23R, which mediates granuloma formation and cell invasion, was identified as one of the multiple differentially methylated gout risk genes. Epigenome-wide analyses revealed differential methylome pathway enrichment for B and T cell receptor signaling, Th17 cell differentiation and interleukin-17 signaling, convergent longevity regulation, circadian entrainment, and AMP-activated protein kinase signaling, which are pathways that impact inflammation via insulin-like growth factor 1 receptor, phosphatidylinositol 3-kinase/Akt, NF-κB, mechanistic target of rapamycin signaling, and autophagy. The gout cohorts overlapped for 37 (52.9%) of the 70 TFs with hypomethylated sequence enrichment and for 30 (78.9%) of the 38 enriched KEGG pathways identified via TFs. Evidence of shared differentially methylated gout TF-gene networks, including the NF-κB activation-limiting TFs MEF2C and NFATC2, pointed to osteoclast differentiation as the most strongly weighted differentially methylated pathway that overlapped in both gout cohorts. CONCLUSION These findings of differential DNA methylation of networked signaling, transcriptional, innate and adaptive immunity, and osteoclastogenesis genes and pathways suggest that they could serve as novel therapeutic targets in the management of flares, tophi, chronic synovitis, and bone erosion in patients with gout.
Collapse
Affiliation(s)
| | | | | | - Ru Liu-Bryan
- University of California, San Diego and San Diego VAMC
| | | | | | - Jun Wang
- University of California, San Diego
| | | | - Wei Wang
- University of California, San Diego
| | | |
Collapse
|
42
|
Natsume C, Aoki N, Aoyama T, Senda K, Matsui M, Ikegami A, Tanaka K, Azuma YT, Fujita T. Fucoxanthin Ameliorates Atopic Dermatitis Symptoms by Regulating Keratinocytes and Regulatory Innate Lymphoid Cells. Int J Mol Sci 2020; 21:ijms21062180. [PMID: 32235696 PMCID: PMC7139773 DOI: 10.3390/ijms21062180] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Fucoxanthin (FX) is a xanthophyll that is contained abundantly in marine plants. The biological action of FX includes its antioxidant and anti-lipogenic activities, while the precise action of its mechanisms on skin cells has not yet been clarified. The current study examined the effect of FX in comparison with tacrolimus (TAC) on NC/Nga mice, which are an atopic dermatitis (AD) model. FX topical treatment dramatically ameliorated itching behavior over the TAC treatment, which was insufficient for improvement of AD symptoms. In Nc/Nga mice, FX or TAC applied to the skin inhibited eosinophil infiltration with decreased expression of Il-33. FX also stimulated Il-2, Il-5, Il-13, Il-10, and TGF-β expression levels, and Sca1+Il-10+TGF-β+ regulatory innate lymphoid cells (ILCreg) were dominantly observed in FX treated skin epidermal keratinocytes and dermal layers. This combined evidence demonstrated that FX exerts anti-inflammatory effects on keratinocytes and ameliorates AD symptoms by regulating ILCreg to normalize immune responses in an atopic dermatitis model.
Collapse
Affiliation(s)
- Chika Natsume
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Nao Aoki
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Tomoko Aoyama
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Keisuke Senda
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Mio Matsui
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Airi Ikegami
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Kosuke Tanaka
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University, Graduate School of Life and Environmental Science, Izumisano, Osaka 598-8531, Japan;
| | - Takashi Fujita
- Molecular Toxicology Lab., Ritsumeikan University, Shiga 525-8577, Japan; (C.N.); (N.A.); (T.A.); (K.S.); (M.M.); (A.I.); (K.T.)
- Correspondence: ; Tel.: +81-77-561-2848
| |
Collapse
|
43
|
Schein CH. Repurposing approved drugs on the pathway to novel therapies. Med Res Rev 2020; 40:586-605. [PMID: 31432544 PMCID: PMC7018532 DOI: 10.1002/med.21627] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
The time and cost of developing new drugs have led many groups to limit their search for therapeutics to compounds that have previously been approved for human use. Many "repurposed" drugs, such as derivatives of thalidomide, antibiotics, and antivirals have had clinical success in treatment areas well beyond their original approved use. These include applications in treating antibiotic-resistant organisms, viruses, cancers and to prevent burn scarring. The major theoretical justification for reusing approved drugs is that they have known modes of action and controllable side effects. Coadministering antibiotics with inhibitors of bacterial toxins or enzymes that mediate multidrug resistance can greatly enhance their activity. Drugs that control host cell pathways, including inflammation, tumor necrosis factor, interferons, and autophagy, can reduce the "cytokine storm" response to injury, control infection, and aid in cancer therapy. An active compound, even if previously approved for human use, will be a poor clinical candidate if it lacks specificity for the new target, has poor solubility or can cause serious side effects. Synergistic combinations can reduce the dosages of the individual components to lower reactivity. Preclinical analysis should take into account that severely ill patients with comorbidities will be more sensitive to side effects than healthy trial subjects. Once an active, approved drug has been identified, collaboration with medicinal chemists can aid in finding derivatives with better physicochemical properties, specificity, and efficacy, to provide novel therapies for cancers, emerging and rare diseases.
Collapse
Affiliation(s)
- Catherine H Schein
- Department of Biochemistry and Molecular Biology, Institute for Human Infection and Immunity (IHII), University of Texas Medical Branch at Galveston, Galveston, Texas
| |
Collapse
|
44
|
Mitochondrial permeability transition pore is involved in oxidative burst and NETosis of human neutrophils. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165664. [PMID: 31926265 DOI: 10.1016/j.bbadis.2020.165664] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/13/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
Neutrophils release neutrophil extracellular traps (NETs) in response to numerous pathogenic microbes as the last suicidal resource (NETosis) in the fight against infection. Apart from the host defense function, NETs play an essential role in the pathogenesis of various autoimmune and inflammatory diseases. Therefore, understanding the molecular mechanisms of NETosis is important for regulating aberrant NET release. The initiation of NETosis after the recognition of pathogens by specific receptors is mediated by an increase in intracellular Ca2+ concentration, therefore, the use of Ca2+ ionophore A23187 can be considered a semi-physiological model of NETosis. Induction of NETosis by various stimuli depends on reactive oxygen species (ROS) produced by NADPH oxidase, however, NETosis induced by Ca2+ ionophores was suggested to be mediated by ROS produced in mitochondria (mtROS). Using the mitochondria-targeted antioxidant SkQ1 and specific inhibitors of NADPH oxidase, we showed that both sources of ROS, mitochondria and NADPH oxidase, are involved in NETosis induced by A23187 in human neutrophils. In support of the critical role of mtROS, SkQ1-sensitive NETosis was demonstrated to be induced by A23187 in neutrophils from patients with chronic granulomatous disease (CGD). We assume that Ca2+-triggered mtROS production contributes to NETosis either directly (CGD neutrophils) or by stimulating NADPH oxidase. The opening of the mitochondrial permeability transition pore (mPTP) in neutrophils treated by A23187 was revealed using the electron transmission microscopy as a swelling of the mitochondrial matrix. Using specific inhibitors, we demonstrated that the mPTP is involved in mtROS production, NETosis, and the oxidative burst induced by A23187.
Collapse
|
45
|
Zhang L, He Y, Wu C, Wu M, Chen X, Luo J, Cai Y, Xia P, Chen B. Altered expression of glucose metabolism associated genes in a tacrolimus‑induced post‑transplantation diabetes mellitus in rat model. Int J Mol Med 2019; 44:1495-1504. [PMID: 31432104 DOI: 10.3892/ijmm.2019.4313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 04/17/2019] [Indexed: 11/06/2022] Open
Abstract
Post‑transplantation diabetes mellitus (PTDM) is a known side effect in transplant recipients administered with immunosuppressant drugs, such as tacrolimus (Tac). Although injury of islet cells is considered a major reason for Tac‑induced PTDM, the involvement of insulin resistance in PTDM remains unknown. In the present study, expression levels of adipocytokines, glucose metabolism associated genes and peroxisome proliferator‑activated receptor (PPAR)‑γ in adipose, muscular and liver tissues from a rat model induced with Tac (1 mg/kg/day) were examined. Rats developed hyperglycemia and glucose intolerance after 10 days of Tac administration. A subgroup of diabetic rats was further treated with rosiglitazone (4 mg/kg), a PPAR‑γ activator. Adipose, muscle and liver tissues were obtained on day 15 after induction and the results demonstrated that expression levels of adipocytokines, PPAR‑γ and proteins in the insulin associated signaling pathway varied in the different groups. Rosiglitazone administration significantly improved hyperglycemia, glucose intolerance and expression levels of proteins associated with insulin signaling, as well as adipocytokines expression. The results of this study demonstrated that adipocytokines and PPAR‑γ signaling may serve important roles in the pathogenesis of Tac‑induced PTDM, which may provide a promising application in the treatment of PTDM in the future.
Collapse
Affiliation(s)
- Ling Zhang
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yunqiang He
- Department of Endocrinology and Metabolism, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Cunzao Wu
- Department of Transplantation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Minmin Wu
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xuehai Chen
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jiao Luo
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yong Cai
- Department of Transplantation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Peng Xia
- Department of Transplantation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Key Laboratory of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
46
|
Heizmann CW. Ca 2+-Binding Proteins of the EF-Hand Superfamily: Diagnostic and Prognostic Biomarkers and Novel Therapeutic Targets. Methods Mol Biol 2019; 1929:157-186. [PMID: 30710273 DOI: 10.1007/978-1-4939-9030-6_11] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A multitude of Ca2+-sensor proteins containing the specific Ca2+-binding motif (helix-loop-helix, called EF-hand) are of major clinical relevance in a many human diseases. Measurements of troponin, the first intracellular Ca-sensor protein to be discovered, is nowadays the "gold standard" in the diagnosis of patients with acute coronary syndrome (ACS). Mutations have been identified in calmodulin and linked to inherited ventricular tachycardia and in patients affected by severe cardiac arrhythmias. Parvalbumin, when introduced into the diseased heart by gene therapy to increase contraction and relaxation speed, is considered to be a novel therapeutic strategy to combat heart failure. S100 proteins, the largest subgroup with the EF-hand protein family, are closely associated with cardiovascular diseases, various types of cancer, inflammation, and autoimmune pathologies. The intention of this review is to summarize the clinical importance of this protein family and their use as biomarkers and potential drug targets, which could help to improve the diagnosis of human diseases and identification of more selective therapeutic interventions.
Collapse
Affiliation(s)
- Claus W Heizmann
- Department of Pediatrics, Division of Clinical Chemistry and Biochemistry, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
47
|
Durand N, Russell A, Zubair AC. Effect of Comedications and Endotoxins on Mesenchymal Stem Cell Secretomes, Migratory and Immunomodulatory Capacity. J Clin Med 2019; 8:jcm8040497. [PMID: 30979082 PMCID: PMC6517980 DOI: 10.3390/jcm8040497] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are becoming an increasingly popular therapeutic option among patients with a broad range of ailments to modulate immunity and induce regeneration. The majority of patients receiving these MSC therapies are on concurrent medication or have ongoing infection. In the present study, we examined the effect of immunosuppressive drugs and lipopolysaccharides (LPS)/endotoxins on the secretory profile, migration towards site of injury, and suppression of lymphocyte proliferation of bone marrow-derived MSCs (BMSCs). Generally, LPS coculture augmented the secretory capacity of BMSCs while exposure to immunosuppressive drugs resulted primarily in no change or attenuated secretion, with some cases of increased secretion, dependent on the cytokine assayed. Among the immunosuppressants evaluated, Hydrocortisone had the most widespread inhibitory effect, while LPS from E. coli O111:B4 had the most potent stimulatory effect. In addition, we also showed that Hydrocortisone or LPS from E. coli O111:B4 affected the migratory and immunosuppressive capacity of BMSCs. Following simulation with Hydrocortisone, BMSC migration was attenuated, and immunosuppressive capacity against T cell proliferation was enhanced, however, the opposite effects were seen with LPS from E. coli O111:B4. Our data suggests that the clinical outcomes of MSC-based therapy are affected by the use of immunosuppressive medication or the presence of endotoxemia in patients.
Collapse
Affiliation(s)
- Nisha Durand
- Transfusion Medicine, Department of Laboratory Medicine and Pathology and Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | | | |
Collapse
|
48
|
Bendíčková K, Tidu F, De Zuani M, Kohoutková MH, Andrejčinová I, Pompeiano A, Bělášková S, Forte G, Zelante T, Frič J. Calcineurin inhibitors reduce NFAT-dependent expression of antifungal pentraxin-3 by human monocytes. J Leukoc Biol 2019; 107:497-508. [PMID: 30934147 PMCID: PMC7064969 DOI: 10.1002/jlb.4vma0318-138r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 02/20/2019] [Accepted: 03/22/2019] [Indexed: 01/14/2023] Open
Abstract
Calcineurin (CN) inhibitors are effective clinical immunosuppressants but leave patients vulnerable to potentially fatal fungal infections. This study tested the hypothesis that CN inhibition interferes with antifungal immune defenses mediated by monocytes. We showed that NFAT is expressed by human monocytes, and is activated by exposure to fungal ligands. We confirmed that NFAT translocation potently activated target gene transcription using a human monocytic reporter cell line. Inhibition of CN‐NFAT by cyclosporine A significantly reduced monocyte production of TNF‐α, IL‐10, and MCP‐1 proteins in response to pattern recognition receptor ligands as well as to Aspergillus fumigatus conidia. Moreover, we revealed that human monocytes express the antifungal protein pentraxin‐3 under control of NFAT. In conclusion, clinical CN inhibitors have the potential to interfere with the novel NFAT‐dependent pentraxin‐3 pathway as well as antifungal cytokine production in human monocytes, thereby impeding monocyte‐mediated defenses against fungal infection in immune‐suppressed patients.
Collapse
Affiliation(s)
- Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Federico Tidu
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marco De Zuani
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | | | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Antonio Pompeiano
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Silvie Bělášková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Giancarlo Forte
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
49
|
Zmonarski SC, Madziarska K, Golebiowski T, Banasik M, Mazanowska O, Madziarski M, Krajewska M. Can the Toll-like receptors 4 expression in peripheral blood mononuclear cells help assess the effectiveness of immunosuppression and the chance of a future good renal transplant function? Transpl Immunol 2018; 53:43-50. [PMID: 30592973 DOI: 10.1016/j.trim.2018.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 12/24/2018] [Accepted: 12/24/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND A small percentage of peripheral blood mononuclear cells (PBMCs) circulating during the kidney transplantation (KT) period remain in the blood long after transplantation. A part of the PBMCs penetrates the graft. AIM To examine if the choice of immunosuppression may change TLR4ex and how TLR4ex affects the transplant function in the future. MATERIAL The study population-143 transplanted patients (pts) (55 females, 88 males), mean age on recruitment day 50.33 ± 12.8 years old, mean BMI 25.04 ± 4.18. 41 pts. experienced delayed graft function (DGF+). 55 pts. were treated with cyclosporine A (CsA) and 88 with tacrolimus (Tac). All were treated with mofetil mycophenolate (MMF). The PBMCs acquisition and starting point of the follow-up (TLR-day) was at least one month after KT. METHOD We investigated averaged mRNA expression of Toll-like receptors 4 (TLR4ex) in non-stimulated peripheral blood mononuclear cells with the use of real-time polymerase chain reaction. The KT pts. (All, Tac, CsA, DGF+) were divided by the respective median of their TLR4ex (lower: L-TLR4ex, higher: H-TLR4ex). Main clinical parameters and transplant biopsy files (if available) were assessed on TLR-day and post follow-up. RESULTS We found that TLR4ex was reduced for a long time in patients who experienced delayed graft function. L-TLR4ex had a higher proportion of DGF+ patients, and patients treated with CsA but lower of those treated with Tac than in H-TLR4ex. The amplitude of changes in renal function parameters (ΔEGFR%/ΔsCr/ΔsCr%) was clearly less favorable for L-TLR4ex. Tacrolimus expressed a stabilizing effect. Both the positive vasculitis score and chronic graft nephropathy were more frequent in the L-TLR4ex group. On TLR-day an association of renal function and Tac concentration with TLR4ex was clear only in the tacrolimus population. The TLR4ex was lower in patients with a future deterioration of the graft function. CONCLUSION In kidney transplant recipients the occurrence of DGF results in a long-term reduction of the averaged TLR4ex in PBMC. Tacrolimus exerts a clear, stabilizing, positive and dose-dependent effect on TLR4ex. An improvement in renal transplant function may be expected in KT patients with high TLR4ex. Evaluation of the averaged TLR4ex can be used to assess the efficacy of immunosuppression in the treatment with tacrolimus and to estimate the likelihood of deterioration in renal function.
Collapse
Affiliation(s)
- Slawomir C Zmonarski
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| | - Katarzyna Madziarska
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland.
| | - Tomasz Golebiowski
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| | - Miroslaw Banasik
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| | - Oktawia Mazanowska
- Faculty of Medicine and Dentistry, Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| | - Marcin Madziarski
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| | - Magdalena Krajewska
- Dept. of Nephrology and Transplantation Medicine, Medical University Wroclaw, Poland
| |
Collapse
|
50
|
Guo J, Kang P, Zhu L, Sun S, Tao M, Zhang H, Tang B. [Mitochondrial aldehyde dehydrogenase 2 protects against high glucose-induced injury in neonatal rat cardiomyocytes by regulating CaN-NFAT3 signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1288-1293. [PMID: 30514674 DOI: 10.12122/j.issn.1673-4254.2018.11.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate whether CaN-NFAT3 pathway mediates the protective effects of aldehyde dehydrogenase (ALDH) 2 in high glucose-treated neonatal rat ventricular myocytes. METHODS The ventricular myocytes were isolated from the heart of neonatal (within 3 days) SD rats by enzyme digestion and cultured in the presence of 5-Brdu. After reaching confluence, the cultured ventricular myocytes were identified using immunofluorescence assay for α-SA protein. The cells were then cultured in either normal (5 mmol/L) or high glucose (30 mmol/L) medium in the presence of ALDH2 agonist Alda-1, ALDH 2 inhibitor Daidzin, or Alda-1 and NFAT3 inhibitor (11R-VIVIT). Fluorescent probe and ELISA were used to detect intracellular Ca2+ concentration and CaN content, respectively; ALDH2, CaN and NFAT3 protein expressions in the cells were detected using Western blotting. RESULTS Compared with cells cultured in normal glucose, the cells exposed to high glucose showed a significantly decreased expression of ALDH2 protein (P < 0.05) and increased expressions of CaN (P < 0.05) and NFAT3 proteins with also increased intracellular CaN and Ca2+ concentrations (P < 0.01). Alda-1 treatment significantly lowered Ca2+ concentration (P < 0.05), intracellular CaN content (P < 0.01), and CaN and NFAT3 protein expressions (P < 0.05), and increased ALDH2 protein expression (P < 0.05) in high glucose- exposed cells; Daidzin treatment significantly increased Ca2+ concentration (P < 0.01) and intracellular CaN content (P < 0.05) in the exposed cells. Compared with Alda-1 alone, treatment of the high glucose-exposed cells with both Alda-1 and 11R-VIVIT did not produce significant changes in the expression of ALDH2 protein (P>0.05) but significantly reduced the expression of NFAT3 protein (P < 0.05). CONCLUSIONS Mitochondrial ALDH2 protects neonatal rat cardiomyocytes against high glucose-induced injury possibly by negatively regulating Ca2+-CaN-NFAT3 signaling pathway.
Collapse
Affiliation(s)
- Jianlu Guo
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Pinfang Kang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Lei Zhu
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Shuo Sun
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Min Tao
- Department of Cardiology, Huishan District People's Hospital, Wuxi 214100, China
| | - Heng Zhang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Bi Tang
- Department of Cardiology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|