1
|
Wang X, Ni T, Miao J, Huang X, Feng Z. The role and mechanism of triptolide, a potential new DMARD, in the treatment of rheumatoid arthritis. Ageing Res Rev 2025; 104:102643. [PMID: 39722411 DOI: 10.1016/j.arr.2024.102643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Triptolide (TP) is the primary pharmacological component of Tripterygium Glycosides (TG), which has anti-inflammatory, antiproliferative, and immunosuppressive properties, among other pharmacological actions, and has excellent potential for developing into a new DMARD. We have reviewed the effects and mechanisms of TP on immunosuppression, inhibiting synovial proliferation, and preventing articular bone destruction in the treatment of rheumatoid arthritis (RA), which is a common disease in the elderly in this paper. We have found that TP has regulatory effects on multiple vital cells in the above-mentioned pathological process of RA, such as monocytes/macrophages, dendritic cells, T cells, fibroblast-like synoviocytes, and osteoclasts. We also found that TP can regulate multiple key signaling pathways such as NF-κB, JAK/STAT, and MAPK through various molecular regulatory mechanisms, achieving regulatory effects on numerous phenotypes of the above-mentioned vital cells.
Collapse
Affiliation(s)
- Xiwen Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Tianyang Ni
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Jianru Miao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Xinyao Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
2
|
Xiong Y, Yin Y, Darshika Kodithuwakku N, Lv J, Wang J, Ding Y, Chen J. Immunosuppressive effects of triptolide via interleukin-2/receptor signaling. Immunopharmacol Immunotoxicol 2024; 46:727-740. [PMID: 39290043 DOI: 10.1080/08923973.2024.2373219] [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: 03/07/2023] [Accepted: 06/22/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Triptolide (TP) has been confirmed to possess many beneficial functions including anti-inflammation and immunosuppression. OBJECTIVE The present study aimed to explore the potential involvement of IL-2/IL-2R pathway in the immunosuppressive activities of TP. METHODS Cultured CTLL-2 cells were utilized to evaluate the potential benefits of TP. Then cell viability was determined by CCK-8 assay, IFN-γ level by ELISA assay, Annexin V-FITC/PI double-staining and CD25 expression by flow cytometry, and protein expression by western blotting. Additionally, rhIL-2-driven lymphocytes following ConA activation were investigated. The interactions of TP with IL-2 and IL-2Rα were investigated by binding assays and molecular dynamics simulations. RESULTS TP treatment attenuated IFN-γ level and cell viability in both rhIL-2-induced CTLL-2 cells and rhIL-2-driven splenic lymphocytes. TP treatment increased cellular apoptosis/necrosis and cleaved PARP-1 level, while suppressed c-Myc level in rhIL-2-induced CTLL-2 cells. Additionally, TP treatment reduced CD25 expression on CTLL-2 cell surface. Notably, the phosphorylation protein levels in IL-2R signaling pathways were inhibited by TP exposure prior to rhIL-2 stimulation. SPR and BLI assays verified TP that directly bound to rhIL-2 and rmIL-2Rα, respectively. Molecular simulations suggested that TP bound at the interface of IL-2 and IL-2Rα near the hydrophobic patch composed of F62, L92 on IL-2 and L23, I46, V139 on IL-2Rα, resulting in decreased binding free energy between IL-2 and IL-2Rα. CONCLUSIONS These findings collectively emphasized that TP interfered IL-2/IL-2Rα interactions, down-regulated IL-2Rα expression, and inhibited IL-2R signaling pathways activation, thereby leading to the immune cells being desensitized to rhIL-2 and exhibiting immunosuppressive properties.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Pharmacology, Wannan Medical College, Wuhu, China
| | - Yi Yin
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | | | - Jiagang Lv
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Juan Wang
- Department of Pharmacology, Wannan Medical College, Wuhu, China
| | - Yanxia Ding
- Department of Human Anatomy, Wannan Medical College, Wuhu, China
| | - Jiao Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
3
|
Copsel SN, Garrido VT, Barreras H, Bader CS, Pfeiffer B, Mateo-Victoriano B, Wolf D, Gallardo M, Paczesny S, Komanduri KV, Benjamin CL, Villarino AV, Saluja AK, Levy RB. Minnelide suppresses GVHD and enhances survival while maintaining GVT responses. JCI Insight 2024; 9:e165936. [PMID: 38602775 PMCID: PMC11141936 DOI: 10.1172/jci.insight.165936] [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: 10/04/2022] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (aHSCT) can cure patients with otherwise fatal leukemias and lymphomas. However, the benefits of aHSCT are limited by graft-versus-host disease (GVHD). Minnelide, a water-soluble analog of triptolide, has demonstrated potent antiinflammatory and antitumor activity in several preclinical models and has proven both safe and efficacious in clinical trials for advanced gastrointestinal malignancies. Here, we tested the effectiveness of Minnelide in preventing acute GVHD as compared with posttransplant cyclophosphamide (PTCy). Strikingly, we found Minnelide improved survival, weight loss, and clinical scores in an MHC-mismatched model of aHSCT. These benefits were also apparent in minor MHC-matched aHSCT and xenogeneic HSCT models. Minnelide was comparable to PTCy in terms of survival, GVHD clinical score, and colonic length. Notably, in addition to decreased donor T cell infiltration early after aHSCT, several regulatory cell populations, including Tregs, ILC2s, and myeloid-derived stem cells in the colon were increased, which together may account for Minnelide's GVHD suppression after aHSCT. Importantly, Minnelide's GVHD prevention was accompanied by preservation of graft-versus-tumor activity. As Minnelide possesses anti-acute myeloid leukemia (anti-AML) activity and is being applied in clinical trials, together with the present findings, we conclude that this compound might provide a new approach for patients with AML undergoing aHSCT.
Collapse
Affiliation(s)
| | | | | | | | - Brent Pfeiffer
- Department of Pediatrics, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | | | | | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Krishna V. Komanduri
- Department of Microbiology and Immunology
- Sylvester Comprehensive Cancer Center
- Department of Medicine, and
| | - Cara L. Benjamin
- Sylvester Comprehensive Cancer Center
- Department of Medicine, and
| | | | - Ashok K. Saluja
- Department of Surgery, and
- Sylvester Comprehensive Cancer Center
| | - Robert B. Levy
- Department of Microbiology and Immunology
- Sylvester Comprehensive Cancer Center
- Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
4
|
Wang G, Guo H, Ren Y, Chen W, Wang Y, Li J, Liu H, Xing J, Zhang Y, Li N. Triptolide enhances carboplatin-induced apoptosis by inhibiting nucleotide excision repair (NER) activity in melanoma. Front Pharmacol 2023; 14:1157433. [PMID: 37324464 PMCID: PMC10267402 DOI: 10.3389/fphar.2023.1157433] [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: 02/02/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Carboplatin (CBP) is a DNA damaging drug used to treat various cancers, including advanced melanoma. Yet we still face low response rates and short survival due to resistance. Triptolide (TPL) is considered to have multifunctional antitumor effects and has been confirmed to enhance the cytotoxic effects of chemotherapeutic drugs. Herein, we aimed to investigate the knowledge about the effects and mechanisms for the combined application of TPL and CBP against melanoma. Methods: Melanoma cell lines and xenograft mouse model were used to uncover the antitumor effects and the underlying molecular mechanisms of the alone or combined treatment of TPL and CBP in melanoma. Cell viability, migration, invasion, apoptosis, and DNA damage were detected by conventional methods. The rate-limiting proteins of the NER pathway were quantitated using PCR and Western blot. Fluorescent reporter plasmids were used to test the NER repair capacity. Results: Our results showed that the presence of TPL in CBP treatment could selectively inhibit NER pathway activity, and TPL exerts a synergistic effect with CBP to inhibit viability, migration, invasion, and induce apoptosis of A375 and B16 cells. Moreover, combined treatment with TPL and CBP significantly inhibited tumor progression in nude mice by suppressing cell proliferation and inducing apoptosis. Discussion: This study reveals the NER inhibitor TPL which has great potential in treating melanoma, either alone or in combination with CBP.
Collapse
Affiliation(s)
- Geng Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Hongmin Guo
- People’s Hospital of Changshou Chongqing, Chongqing, China
| | - Yan Ren
- Health Science Center, Ningbo University, Ningbo, China
| | - Weiyi Chen
- Health Science Center, Ningbo University, Ningbo, China
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Jianing Li
- Health Science Center, Ningbo University, Ningbo, China
| | - Hua Liu
- Health Science Center, Ningbo University, Ningbo, China
| | - Jingjun Xing
- Health Science Center, Ningbo University, Ningbo, China
| | - Yanru Zhang
- Health Science Center, Ningbo University, Ningbo, China
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Sun L, Chen L, Yang Z, Sun X, Jin D, Qiu Y, Gu W. A novel ratiometric dehydroabietic acid-based fluorescent probe for detecting HPO42- and its application in food samples. J Food Compost Anal 2023. [DOI: 10.1016/j.jfca.2023.105316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
6
|
Mediouni S, Lyu S, Schader SM, Valente ST. Forging a Functional Cure for HIV: Transcription Regulators and Inhibitors. Viruses 2022; 14:1980. [PMID: 36146786 PMCID: PMC9502519 DOI: 10.3390/v14091980] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/28/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Current antiretroviral therapy (ART) increases the survival of HIV-infected individuals, yet it is not curative. The major barrier to finding a definitive cure for HIV is our inability to identify and eliminate long-lived cells containing the dormant provirus, termed viral reservoir. When ART is interrupted, the viral reservoir ensures heterogenous and stochastic HIV viral gene expression, which can reseed infection back to pre-ART levels. While strategies to permanently eradicate the virus have not yet provided significant success, recent work has focused on the management of this residual viral reservoir to effectively limit comorbidities associated with the ongoing viral transcription still observed during suppressive ART, as well as limit the need for daily ART. Our group has been at the forefront of exploring the viability of the block-and-lock remission approach, focused on the long-lasting epigenetic block of viral transcription such that without daily ART, there is no risk of viral rebound, transmission, or progression to AIDS. Numerous studies have reported inhibitors of both viral and host factors required for HIV transcriptional activation. Here, we highlight and review some of the latest HIV transcriptional inhibitor discoveries that may be leveraged for the clinical exploration of block-and-lock and revolutionize the way we treat HIV infections.
Collapse
Affiliation(s)
- Sonia Mediouni
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, 130 Scripps Way, 3C1, Jupiter, FL 33458, USA
| | - Shuang Lyu
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, 130 Scripps Way, 3C1, Jupiter, FL 33458, USA
| | - Susan M. Schader
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA
| | - Susana T. Valente
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, 130 Scripps Way, 3C1, Jupiter, FL 33458, USA
| |
Collapse
|
7
|
Nuzzo G, Senese G, Gallo C, Albiani F, Romano L, d’Ippolito G, Manzo E, Fontana A. Antitumor Potential of Immunomodulatory Natural Products. Mar Drugs 2022; 20:md20060386. [PMID: 35736189 PMCID: PMC9229642 DOI: 10.3390/md20060386] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer is one of the leading causes of death globally. Anticancer drugs aim to block tumor growth by killing cancerous cells in order to prevent tumor progression and metastasis. Efficient anticancer drugs should also minimize general toxicity towards organs and healthy cells. Tumor growth can also be successfully restrained by targeting and modulating immune response. Cancer immunotherapy is assuming a growing relevance in the fight against cancer and has recently aroused much interest for its wider safety and the capability to complement conventional chemotherapeutic approaches. Natural products are a traditional source of molecules with relevant potential in the pharmacological field. The huge structural diversity of metabolites with low molecular weight (small molecules) from terrestrial and marine organisms has provided lead compounds for the discovery of many modern anticancer drugs. Many natural products combine chemo-protective and immunomodulant activity, thus offering the potential to be used alone or in association with conventional cancer therapy. In this review, we report the natural products known to possess antitumor properties by interaction with immune system, as well as discuss the possible immunomodulatory mechanisms of these molecules.
Collapse
Affiliation(s)
- Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
- Correspondence: (G.N.); (E.M.); Tel.: +39-081-8675104 (G.N.); +39-081-8675177 (E.M.)
| | - Giuseppina Senese
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
| | - Carmela Gallo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
| | - Federica Albiani
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
| | - Lucia Romano
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
| | - Giuliana d’Ippolito
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
| | - Emiliano Manzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
- Correspondence: (G.N.); (E.M.); Tel.: +39-081-8675104 (G.N.); +39-081-8675177 (E.M.)
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (G.S.); (C.G.); (F.A.); (L.R.); (G.d.); (A.F.)
- Department of Biology, University of Naples Federico II, Via Cinthia–Bld. 7, 80126 Napoli, Italy
| |
Collapse
|
8
|
Triptolide Suppresses NF-κB-Mediated Inflammatory Responses and Activates Expression of Nrf2-Mediated Antioxidant Genes to Alleviate Caerulein-Induced Acute Pancreatitis. Int J Mol Sci 2022; 23:ijms23031252. [PMID: 35163177 PMCID: PMC8835869 DOI: 10.3390/ijms23031252] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/12/2022] Open
Abstract
Triptolide (TP), the main active ingredient of Tripterygium wilfordii Hook.f., displays potent anti-inflammatory, antioxidant, and antiproliferative activities. In the present study, the effect of TP on acute pancreatitis and the underlying mechanisms of the disease were investigated using a caerulein-induced animal model of acute pancreatitis (AP) and an in vitro cell model. In vivo, pretreatment with TP notably ameliorated pancreatic damage, shown as the improvement in serum amylase and lipase levels and pancreatic morphology. Meanwhile, TP modulated the infiltration of neutrophils and macrophages (Ly6G staining and CD68 staining) and decreased the levels of proinflammatory factors (TNF-α and IL-6) through inhibiting the transactivation of nuclear factor-κB (NF-κB) in caerulein-treated mice. Furthermore, TP reverted changes in oxidative stress markers, including pancreatic glutathione (GSH), superoxide dismutase (SOD), and malondialdehyde (MDA), in acute pancreatitis mice. Additionally, TP pretreatment inhibited intracellular reactive oxygen species (ROS) levels via upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) expression and Nrf2-regulated redox genes expression (HO-1, SOD1, GPx1 and NQO1) in vitro. Taken together, our data suggest that TP exert protection against pancreatic inflammation and tissue damage by inhibiting NF-κB transactivation, modulating immune cell responses and activating the Nrf2-mediated antioxidative system, thereby alleviating acute pancreatitis.
Collapse
|
9
|
König KMK, Jahun AS, Nayak K, Drumright LN, Zilbauer M, Goodfellow I, Hosmillo M. Design, development, and validation of a strand-specific RT-qPCR assay for GI and GII human Noroviruses. Wellcome Open Res 2021; 6:245. [PMID: 34708158 PMCID: PMC8506223 DOI: 10.12688/wellcomeopenres.17078.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2021] [Indexed: 11/20/2022] Open
Abstract
Human noroviruses (HuNoV) are the major cause of viral gastroenteritis worldwide. Similar to other positive-sense single-stranded RNA viruses, norovirus RNA replication requires the formation of a negative strand RNA intermediate. Methods for detecting and quantifying the viral positive or negative sense RNA in infected cells and tissues can be used as important tools in dissecting virus replication. In this study, we have established a sensitive and strand-specific Taqman-based quantitative polymerase chain reaction (qPCR) assay for both genogroups GI and GII HuNoV. This assay shows good reproducibility, has a broad dynamic range and is able to detect a diverse range of isolates. We used tagged primers containing a non-viral sequence for the reverse transcription (RT) reaction and targeted this tag in the succeeding qPCR reaction to achieve strand specificity. The specificity of the assay was confirmed by the detection of specific viral RNA strands in the presence of high levels of the opposing strands, in both RT and qPCR reactions. Finally, we further validated the assay in norovirus replicon-bearing cell lines and norovirus-infected human small intestinal organoids, in the presence or absence of small-molecule inhibitors. Overall, we have established a strand-specific qPCR assay that can be used as a reliable method to understand the molecular details of the human norovirus life cycle.
Collapse
Affiliation(s)
- Katja Marie Kjara König
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
- Insitute of Chemistry and Metabolomics, Center for Structural and Cell Biology in Medicine (CSCM), University of Lübeck, Lübeck, Germany
| | - Aminu S. Jahun
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Komal Nayak
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Lydia N. Drumright
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
10
|
Qiao J, Chen R, Wang M, Bai R, Cui X, Liu Y, Wu C, Chen C. Perturbation of gut microbiota plays an important role in micro/nanoplastics-induced gut barrier dysfunction. NANOSCALE 2021; 13:8806-8816. [PMID: 33904557 DOI: 10.1039/d1nr00038a] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The widespread occurrence of microplastics (MPLs) and nanoplastics (NPLs), collectively abbreviated as M/NPLs, has markedly affected the ecosystem and has become a global threat to human health. Multiple investigations have shown that the chronic ingestion of M/NPLs negatively affects gut barrier function but the mechanism remains unclear. Herein, this research has investigated the toxic effects of pristine polystyrene (PS) M/NPLs, negatively charged carboxylated polystyrene M/NPLs (PS-COOH) and positively charged aminated polystyrene M/NPLs (PS-NH2) of two sizes (70 nm and 5 μm in diameter) in mice. Gavage of these PS M/NPLs for 28 days caused obvious injuries to the gut tract, leading to the decreased expression of tight junction proteins. The toxicity of the M/NPLs was ranked as PS-NH2 > PS-COOH > pristine PS. Oral administration of these M/NPLs resulted in marked gut microbiota dysbiosis. The M/NPLs-enriched genera generally contained opportunistic pathogens which are accompanied by a deteriorated intestinal barrier function, while most M/NPLs-decreased bacteria were beneficial microbes with known tight junction-promoting functions, implicating an important indirect toxic effect of gut microbiota dysbiosis in M/NPLs-induced gut barrier dysfunction. In conclusion, this research highlights the importance of gut microbiota in the toxicity of M/NPLs exposure on gut barrier function, providing novel insights into the adverse effects of M/NPLs exposure on human health.
Collapse
Affiliation(s)
- Jiyan Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China and College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Rui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China
| | - Mengjie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China
| | - Chongming Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience & Technology of China, Beijing, 100190, China and College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
11
|
Zhou P, Guo H, Li Y, Liu Q, Qiao X, Lu Y, Mei PY, Zheng Z, Li J. Monocytes promote pyroptosis of endothelial cells during lung ischemia-reperfusion via IL-1R/NF-κB/NLRP3 signaling. Life Sci 2021; 276:119402. [PMID: 33785335 DOI: 10.1016/j.lfs.2021.119402] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
In our previous study, we observed that donor pulmonary intravascular nonclassical monocytes play a major role in early PGF, but the specific mechanism remained unclear. In this study, we investigated the mechanistic role of monocytes in inducing pyroptosis of human pulmonary microvascular endothelial cells (HPMECs) during IRI. A murine hilar ligation model of IRI was utilized whereby left lungs underwent 1 h of ischemia and 23 h of reperfusion. Monocyte depletion by intraperitoneal clodronate-liposome treatment on pulmonary edema and pyroptosis activation were determined. In vitro experiments, we performed the co-culture experiments under hypoxia-reoxygenation (H/R) conditions to mimic the IRI environment. We monitored the expression of NLRP3, caspase-1 and IL-1β in co-cultures of monocytes (U937 cells) and HPMECs under H/R conditions. NLRP3, IL-1β and IL-1R siRNA knockdown, caspase-1 and NF-κB pathway inhibitors were employed to elucidate the mechanism modulating HPMEC pyroptosis during H/R. Treatment of mice with clodronate-liposome attenuated IR-induced pulmonary edema, cytokine production and pyroptosis activation. In vitro, NLRP3 knockdown in monocytes reduced caspase-1 and IL-1β secretion in co-cultures of monocytes and HPMECs. Reduced HPMEC pyroptosis was also observed either containing HPMECs with genetically engineered IL-1R knockdown or in co-culture treated with a Triplotide inhibitor that disrupts NF-κB signaling. Monocytes play a vital role in the development of transplant-associated ischemia-reperfusion injury. A potential role is that monocytes secrete IL-1β to induce HPMEC pyroptosis via the IL-1R/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Guo
- Department of Thyroid and Breast Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwei Qiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Lu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Yuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhikun Zheng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinsong Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Alimohammadi N, Koosha F, Rafeian-Kopaei M. Current, New and Future Therapeutic Targets in Inflammatory Bowel Disease: A Systematic Review. Curr Pharm Des 2020; 26:2668-2675. [PMID: 32250220 DOI: 10.2174/1381612826666200406081920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/10/2020] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are chronic relapsing conditions resulting from immune system activity in a genetically predisposed individual. IBD is based on progressive damage to the inflamed gut tissue. As its pathogenesis remains unknown, recent accumulating data have demonstrated that IBD is a complex and multi-factorial disorder correlated with host luminal factors, which lead to an imbalance between pro- and anti-inflammatory signaling. The growing understanding of the molecular mechanisms responsible for IBD has suggested a wide range of potential therapeutic targets to treat this condition. Some patients do not have a satisfactory response to current therapeutic medications such as antitumor necrosis factor (TNF) agents, or their response decreases over time. As a result, IBD therapeutics have been changed recently, with several new agents being evaluated. The identification of various inflammatory cascades has led to forming the idea to have novel medications developed. Medications targeting Janus kinases (JAK), leukocyte trafficking Interleukin (IL) 12/23, and Sphingosine 1 phosphate (S1P) are among these newly developed medications and highlight the role of microbial-host interaction in inflammation as a safe promising strategy. This systematic review aims to summarize different molecular targeting therapeutics, the most potent candidates for IBD treatment in recent studies.
Collapse
Affiliation(s)
- Niloufar Alimohammadi
- Department of Medicine, New York University School of Medicine, New York, New York, United States
| | - Farzad Koosha
- Department of Oral Biology and Pathology, School of Dental Medicine, State University of New York at Stony Brook, New York, United States
| | - Mahmoud Rafeian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahre-kord University of Medical Sciences, Shahre-kord, Iran
| |
Collapse
|
13
|
Vinogradova EV, Zhang X, Remillard D, Lazar DC, Suciu RM, Wang Y, Bianco G, Yamashita Y, Crowley VM, Schafroth MA, Yokoyama M, Konrad DB, Lum KM, Simon GM, Kemper EK, Lazear MR, Yin S, Blewett MM, Dix MM, Nguyen N, Shokhirev MN, Chin EN, Lairson LL, Melillo B, Schreiber SL, Forli S, Teijaro JR, Cravatt BF. An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells. Cell 2020; 182:1009-1026.e29. [PMID: 32730809 PMCID: PMC7775622 DOI: 10.1016/j.cell.2020.07.001] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/14/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
Electrophilic compounds originating from nature or chemical synthesis have profound effects on immune cells. These compounds are thought to act by cysteine modification to alter the functions of immune-relevant proteins; however, our understanding of electrophile-sensitive cysteines in the human immune proteome remains limited. Here, we present a global map of cysteines in primary human T cells that are susceptible to covalent modification by electrophilic small molecules. More than 3,000 covalently liganded cysteines were found on functionally and structurally diverse proteins, including many that play fundamental roles in immunology. We further show that electrophilic compounds can impair T cell activation by distinct mechanisms involving the direct functional perturbation and/or degradation of proteins. Our findings reveal a rich content of ligandable cysteines in human T cells and point to electrophilic small molecules as a fertile source for chemical probes and ultimately therapeutics that modulate immunological processes and their associated disorders.
Collapse
Affiliation(s)
| | - Xiaoyu Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David Remillard
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel C Lazar
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Radu M Suciu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yujia Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Giulia Bianco
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Yamashita
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Medicinal Chemistry Research Laboratories, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd., 463-10 Kawauchi-cho, Tokushima 771-0192, Japan
| | - Vincent M Crowley
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael A Schafroth
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Minoru Yokoyama
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David B Konrad
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth M Lum
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel M Simon
- Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA
| | - Esther K Kemper
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael R Lazear
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sifei Yin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Megan M Blewett
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Melissa M Dix
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nhan Nguyen
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emily N Chin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John R Teijaro
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
14
|
Campos ACP, Antunes GF, Matsumoto M, Pagano RL, Martinez RCR. Neuroinflammation, Pain and Depression: An Overview of the Main Findings. Front Psychol 2020; 11:1825. [PMID: 32849076 PMCID: PMC7412934 DOI: 10.3389/fpsyg.2020.01825] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic pain is a serious public health problem with a strong affective-motivational component that makes it difficult to treat. Most patients with chronic pain suffer from severe depression; hence, both conditions coexist and exacerbate one another. Brain inflammatory mediators are critical for maintaining depression-pain syndrome and could be substrates for it. The goal of our paper was to review clinical and preclinical findings to identify the neuroinflammatory profile associated with the cooccurrence of pain and depression. In addition, we aimed to explore the regulatory effect of neuronal reorganization on the inflammatory response in pain and depression. We conducted a quantitative review supplemented by manual screening. Our results revealed inflammatory signatures in different preclinical models and clinical articles regarding depression-pain syndrome. We also identified that improvements in depressive symptoms and amelioration of pain can be modulated through direct targeting of inflammatory mediators, such as cytokines and molecular inhibitors of the inflammatory cascade. Additionally, therapeutic targets that improve and regulate the synaptic environment and its neurotransmitters may act as anti-inflammatory compounds, reducing local damage-associated molecular patterns and inhibiting the activation of immune and glial cells. Taken together, our data will help to better elucidate the neuroinflammatory profile in pain and depression and may help to identify pharmacological targets for effective management of depression-pain syndrome.
Collapse
Affiliation(s)
| | | | - Marcio Matsumoto
- Anesthesiology Medical Center, Hospital Sirio-Libanes, São Paulo, Brazil
| | | | - Raquel Chacon Ruiz Martinez
- Division of Neuroscience, Hospital Sirio-Libanes, São Paulo, Brazil.,LIM 23, Institute of Psychiatry, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
15
|
Li A, Yang Y, Wang W, Liu Q, Sun Y, Gu W. Synthesis, cytotoxicity and apoptosis‐inducing activity of novel
1
H
‐benzo[
d
]imidazole derivatives of dehydroabietic acid. J CHIN CHEM SOC-TAIP 2020. [DOI: 10.1002/jccs.202000075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- A‐Liang Li
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| | - Ya‐Qun Yang
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| | - Wen‐Yan Wang
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| | - Qing‐Song Liu
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| | - Yue Sun
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| | - Wen Gu
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro‐forest Biomass, Jiangsu Key Lab of Biomass‐based Green Fuels and Chemicals, Co‐Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering Nanjing Forestry University Nanjing P. R. China
| |
Collapse
|
16
|
Li EQ, Zhang JL. Therapeutic effects of triptolide from Tripterygium wilfordii Hook. f. on interleukin-1-beta-induced osteoarthritis in rats. Eur J Pharmacol 2020; 883:173341. [PMID: 32634440 DOI: 10.1016/j.ejphar.2020.173341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 01/25/2023]
Abstract
Osteoarthritis (OA) is a common yet destructive disease affecting the articular cartilage, and is a major cause of immense suffering and disability for millions of people. Previous studies have shown that triptolide (TPL), an active compound derived from Tripterygium wilfordii, has potent immunosuppressive and anti-inflammatory activities useful for treating chronic diseases. However, whether TPL has immunosuppressive activity against OA is not known. In this study, we assessed the therapeutic effects of TPL on interleukin-1-beta (IL-1β)-induced OA in rats. Histological and protein analyses revealed that TPL not only could inhibit interleukin-6 (IL-6) and cyclooxygenase-2 (COX2) protein expression in cells and disrupt inflammation, but it also reduced the expression of matrix metalloproteinase (MMP)-3 and 13. Our results also supported the ability of TPL to suppress the osteoprotegerin/receptor activator of nuclear factor kappa-beta (NF-κB)/receptor activator of NF-κB ligand (OPG/RANK/RANKL) and NF-κB signaling pathways induced by IL-1β. Together these data suggest that TPL may be a potentially valuable treatment for OA, regulating associated inflammation and pain.
Collapse
Affiliation(s)
- En-Qi Li
- Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China
| | - Jin-Li Zhang
- Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China.
| |
Collapse
|
17
|
Yang YQ, Chen H, Liu QS, Sun Y, Gu W. Synthesis and anticancer evaluation of novel 1H-benzo[d]imidazole derivatives of dehydroabietic acid as PI3Kα inhibitors. Bioorg Chem 2020; 100:103845. [PMID: 32344183 DOI: 10.1016/j.bioorg.2020.103845] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Phosphatidylinositol 3-kinase (PI3K) is one of the most attractive therapeutic targets for cancer treatment. In this study, a series of new 2-arylthio- and 2-arylamino-1H-benzo[d]imidazole derivatives of dehydroabietic acid were designed, synthesized and characterized by 1H NMR, 13C NMR, IR and MS spectra analyses. In the in vitro anticancer assay, some title compounds showed significant inhibitory activities against four cancer cell lines (HCT-116, MCF-7, HeLa and HepG2). Among them, compound 9g exhibited the most potent activity with IC50 values of 0.18 ± 0.03, 0.43 ± 0.05, 0.71 ± 0.08 and 0.63 ± 0.09 μM against four cancer cell lines, and considerably lower cytotoxicity to human gastric mucosal cell line Ges-1 (IC50: 21.95 ± 0.73 μM). Besides, compound 9g displayed a certain selective activity to PI3Kα (IC50 = 0.012 ± 0.002 μM) over PI3Kβ, γ and δ, and meanwhile, it can remarkably decrease the expression level of p-Akt (Ser473). In addition, compound 9g could increase intracellular reactive oxygen species level, decrease mitochondrial membrane potential, upregulate Bax and cleaved caspase-3/9 levels, downregulate Bcl-2 level and thus induce the apoptosis of HCT-116 cells in a dose-dependent manner. The results suggested that compound 9g could be considered as a promising PI3Kα inhibitor.
Collapse
Affiliation(s)
- Ya-Qun Yang
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Co-Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Hao Chen
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Co-Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Qing-Song Liu
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Co-Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Yue Sun
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Co-Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Wen Gu
- Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Co-Inovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, PR China.
| |
Collapse
|
18
|
Jin H, Li D, Lin MH, Li L, Harrich D. Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure. Viruses 2020; 12:v12040415. [PMID: 32276443 PMCID: PMC7232260 DOI: 10.3390/v12040415] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV) establishes a chronic infection that can be well controlled, but not cured, by combined antiretroviral therapy (cART). Interventions have been explored to accomplish a functional cure, meaning that a patient remains infected but HIV is undetectable in the blood, with the aim of allowing patients to live without cART. Tat, the viral transactivator of transcription protein, plays a critical role in controlling HIV transcription, latency, and viral rebound following the interruption of cART treatment. Therefore, a logical approach for controlling HIV would be to block Tat. Tackling Tat with inhibitors has been a difficult task, but some recent discoveries hold promise. Two anti-HIV proteins, Nullbasic (a mutant of Tat) and HT1 (a fusion of HEXIM1 and Tat functional domains) inhibit viral transcription by interfering with the interaction of Tat and cellular factors. Two small molecules, didehydro-cortistatin A (dCA) and triptolide, inhibit Tat by different mechanisms: dCA through direct binding and triptolide through enhanced proteasomal degradation. Finally, two Tat-based vaccines under development elicit Tat-neutralizing antibodies. These vaccines have increased the levels of CD4+ cells and reduced viral loads in HIV-infected people, suggesting that the new vaccines are therapeutic. This review summarizes recent developments of anti-Tat agents and how they could contribute to a functional cure for HIV.
Collapse
Affiliation(s)
- Hongping Jin
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Dongsheng Li
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Min-Hsuan Lin
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia;
| | - David Harrich
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
- Correspondence: ; Tel.: +617-3845-3679
| |
Collapse
|
19
|
Hosmillo M, Chaudhry Y, Nayak K, Sorgeloos F, Koo BK, Merenda A, Lillestol R, Drumright L, Zilbauer M, Goodfellow I. Norovirus Replication in Human Intestinal Epithelial Cells Is Restricted by the Interferon-Induced JAK/STAT Signaling Pathway and RNA Polymerase II-Mediated Transcriptional Responses. mBio 2020; 11:e00215-20. [PMID: 32184238 PMCID: PMC7078467 DOI: 10.1128/mbio.00215-20] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Human noroviruses (HuNoV) are a leading cause of viral gastroenteritis worldwide and a significant cause of morbidity and mortality in all age groups. The recent finding that HuNoV can be propagated in B cells and mucosa-derived intestinal epithelial organoids (IEOs) has transformed our ability to dissect the life cycle of noroviruses. Using transcriptome sequencing (RNA-Seq) of HuNoV-infected intestinal epithelial cells (IECs), we have found that replication of HuNoV in IECs results in interferon (IFN)-induced transcriptional responses and that HuNoV replication in IECs is sensitive to IFN. This contrasts with previous studies that suggested that the innate immune response may play no role in the restriction of HuNoV replication in immortalized cells. We demonstrated that inhibition of Janus kinase 1 (JAK1)/JAK2 enhanced HuNoV replication in IECs. Surprisingly, targeted inhibition of cellular RNA polymerase II-mediated transcription was not detrimental to HuNoV replication but instead enhanced replication to a greater degree than blocking of JAK signaling directly. Furthermore, we demonstrated for the first time that IECs generated from genetically modified intestinal organoids, engineered to be deficient in the interferon response, were more permissive to HuNoV infection. Taking the results together, our work revealed that IFN-induced transcriptional responses restrict HuNoV replication in IECs and demonstrated that inhibition of these responses mediated by modifications of the culture conditions can greatly enhance the robustness of the norovirus culture system.IMPORTANCE Noroviruses are a major cause of gastroenteritis worldwide, and yet the challenges associated with their growth in culture have greatly hampered the development of therapeutic approaches and have limited our understanding of the cellular pathways that control infection. Here, we show that human intestinal epithelial cells, which represent the first point of entry of human noroviruses into the host, limit virus replication by induction of innate responses. Furthermore, we show that modulating the ability of intestinal epithelial cells to induce transcriptional responses to HuNoV infection can significantly enhance human norovirus replication in culture. Collectively, our findings provide new insights into the biological pathways that control norovirus infection but also identify mechanisms that enhance the robustness of norovirus culture.
Collapse
Affiliation(s)
- Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Komal Nayak
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Bon-Kyoung Koo
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Alessandra Merenda
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Reidun Lillestol
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Lydia Drumright
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Matthias Zilbauer
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
Burki S, Noda K, Philips BJ, Velayutham M, Shiva S, Sanchez PG, Kumar A, D'Cunha J. Impact of triptolide during ex vivo lung perfusion on grafts after transplantation in a rat model. J Thorac Cardiovasc Surg 2020; 161:S0022-5223(20)30191-4. [PMID: 32169373 DOI: 10.1016/j.jtcvs.2019.12.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/30/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Ex vivo lung perfusion creates a proinflammatory environment leading to deterioration in graft quality that may contribute to post-transplant graft dysfunction. Triptolide has been shown to have a therapeutic potential in various disease states because of its anti-inflammatory properties. On this basis, we investigated the impact of triptolide on graft preservation during ex vivo lung perfusion and associated post-transplant outcomes in a rat transplant model. METHODS We performed rat normothermic ex vivo lung perfusion with acellular Steen solution containing 100 nM triptolide for 4 hours and compared the data with untreated lungs. Orthotopic single lung transplantation after ex vivo lung perfusion was performed. RESULTS Physiologic and functional parameters of lung grafts on ex vivo lung perfusion with triptolide were better than those without treatment. Graft glucose consumption was significantly attenuated on ex vivo lung perfusion with triptolide via inhibition of hypoxia signaling resulting in improved mitochondrial function and reduced oxidative stress. Also, intragraft inflammation was markedly lower in triptolide-treated lungs because of inhibition of nuclear factor-κB signaling. Furthermore, post-transplant graft function and inflammatory events were significantly improved in the triptolide group compared with the untreated group. CONCLUSIONS Treatment of lung grafts with triptolide during ex vivo lung perfusion may serve to enhance graft preservation and improve graft protection resulting in better post-transplant outcomes.
Collapse
Affiliation(s)
- Sarah Burki
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Brian J Philips
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Murugesan Velayutham
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pa; Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Sruti Shiva
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Pablo G Sanchez
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Ajay Kumar
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Pheonix, Ariz.
| |
Collapse
|
21
|
Jian C, Zhang L, Jinlong L, Bo T, Liu Z. Effects of brazilein on PSD-95 protein expression and neurological recovery in mice after sciatic nerve injury. Neurosci Lett 2020; 715:134547. [PMID: 31629776 DOI: 10.1016/j.neulet.2019.134547] [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: 07/31/2018] [Revised: 08/23/2019] [Accepted: 10/09/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE To evaluate the local nerve myelin recovery and the expression of PSD-95 protein and mRNA in the L4-L6 segment of the spinal cord after applying Brazilein to sciatic nerve injury BALB/c mice model and investigate the regulatory effects of Brazilein on myelin recovery after peripheral nerve injury. METHODS A total of 160 BALB/c mice were selected to establish the unilateral sciatic nerve injury model and randomly divided into four groups: saline blank control, Brazilein high-dose, medium-dose, and low-dose. Mice were assessed at different time points (1 w, 2 w, 4 w, 8 w) after sciatic nerve injury for the sciatic functional index (SFI) and sciatic nerve function recovery of the injured side by myelin Luxol Fast Blue (LFB) staining of the sciatic nerve. In addition, immunohistochemistry, real time-PCR, and Western blot were used to detect the PSD-95 expression in the spinal cord L4-L6 segments of the injured sciatic nerve at each time point. RESULTS The results of SFI and sciatic nerve function recovery, as well as, myelin LFB staining of the injured side indicated that all indexes of the Brazilein middle- and high-dose groups were significantly better than the low-dose and blank control groups at each time point. The PSD-95 expression in the L4-L6 segment of the spinal cord was statistically lower in the high- and medium-dose groups than in the low-dose and blank control groups at 1 w, 2 w, and 4 w, while the differences between the groups were not significant at 8 w. CONCLUSION Brazilein inhibits PSD-95 activation in the corresponding segment of sciatic nerve spinal cord in BALB/c mice after sciatic nerve injury, thereby inhibiting the excessive expression of free radicals and promoting myelin regeneration.
Collapse
Affiliation(s)
- Cao Jian
- Department of Orthopedic, Affiliated Hospital of Chifeng University, Chifeng 024000, China.
| | - Limin Zhang
- Department of Orthopedic, Affiliated Hospital of Chifeng University, Chifeng 024000, China.
| | - Li Jinlong
- Department of Ophthalmology, Affiliated Hospital of Chifeng University, Chifeng 024000, China.
| | - Tao Bo
- Department of Emergency, Affiliated Hospital of Chifeng University, Chifeng 024000, China.
| | - Zhongxing Liu
- Department of Orthopedic, Affiliated Hospital of Chifeng University, Chifeng 024000, China.
| |
Collapse
|
22
|
Yuan K, Li X, Lu Q, Zhu Q, Jiang H, Wang T, Huang G, Xu A. Application and Mechanisms of Triptolide in the Treatment of Inflammatory Diseases-A Review. Front Pharmacol 2019; 10:1469. [PMID: 31866868 PMCID: PMC6908995 DOI: 10.3389/fphar.2019.01469] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022] Open
Abstract
Bioactive compounds from medicinal plants with anti-inflammatory and immunosuppressive effects have been emerging as important sources of drugs for the treatment of inflammatory disorders. Triptolide, a diterpene triepoxide, is a pharmacologically active compound isolated from Tripterygium wilfordii Hook F (TwHF) that is used as a remedy for inflammatory and autoimmune diseases. As the most promising bioactive compound obtained from TwHF, triptolide has attracted considerable interest recently, especially for its potent anti-inflammatory and immunosuppressive activities. Over the past few years, an increasing number of studies have been published emphasizing the value of triptolide in the treatment of diverse inflammatory disorders. Here, we systematically review the mechanism of action and the therapeutic properties of triptolide in various inflammatory diseases according to different systematic organs, including lupus nephritis, inflammatory bowel disease, asthma, and rheumatoid arthritis with pubmed and Embase. Based on this review, potential research strategies might contribute to the clinical application of triptolide in the future.
Collapse
Affiliation(s)
- Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qingqing Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,State Key Laboratory of Biocontrol, Department of Biochemistry, School of Life Sciences, Sun Yat-Sen (Zhongshan) University, Guangzhou, China
| |
Collapse
|
23
|
GYY4137 Attenuates Sodium Deoxycholate-Induced Intestinal Barrier Injury Both In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5752323. [PMID: 31737669 PMCID: PMC6815576 DOI: 10.1155/2019/5752323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/10/2019] [Indexed: 11/17/2022]
Abstract
Objectives Substantial studies have demonstrated that an elevated concentration of deoxycholic acid (DCA) in the colonic lumen may play a critical role in the pathogenesis of intestinal barrier dysfunction and inflammatory bowel disease (IBD). The purpose of this study was to investigate the protective effects of GYY4137, as a novel and synthetic H2S donor, on the injury of intestinal barrier induced by sodium deoxycholate (SDC) both in vivo and in vitro. Methods In this study, Caco-2 monolayers and mouse models with high SDC concentration in the lumen were used to study the effect of GYY4137 on intestinal barrier dysfunction induced by SDC and its underlying mechanisms. Results In Caco-2 monolayers, a short period of addition of SDC increased the permeability of monolayers obviously, changed distribution of tight junctions (TJs), and improved the phosphorylation level of myosin light chain kinase (MLCK) and myosin light chain (MLC). However, pretreatment with GYY4137 markedly ameliorated the SDC-induced barrier dysfunction. Being injected with GYY4137 could enable mice to resist the SDC-induced injury of the intestinal barrier. Besides, GYY4137 promoted the recovery of the body weight and intestinal barrier histological score of mice with the gavage of SDC. GYY4137 also attenuated the decreased expression level of TJs in mice treated with SDC. Conclusion Taken together, this research suggests that GYY4137 preserves the intestinal barrier from SDC-induced injury via suppressing the activation of P-MLCK-P-MLC2 signaling pathway and increasing the expression level of tight junctions.
Collapse
|
24
|
Hou W, Liu B, Xu H. Triptolide: Medicinal chemistry, chemical biology and clinical progress. Eur J Med Chem 2019; 176:378-392. [DOI: 10.1016/j.ejmech.2019.05.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/11/2019] [Accepted: 05/11/2019] [Indexed: 12/14/2022]
|
25
|
Tomioka H, Tatano Y, Shimizu T, Sano C. Clinical and Basic Studies on Therapeutic Efficacy of Herbal Medicines against Mycobacterial Infections. MEDICINES 2019; 6:medicines6020067. [PMID: 31248144 PMCID: PMC6630501 DOI: 10.3390/medicines6020067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
The high incidence of tuberculosis (TB) in developing countries, the resurgence of TB in industrialized countries, and the worldwide increase in the prevalence of Mycobacterium avium complex infections are important global health concerns. However, the development of novel antimycobacterial drugs is currently making very slow progress. Therefore, it is considered that devising improved administration protocols for clinical treatment against intractable mycobacteriosis using existing chemotherapeutics is more practical than awaiting the development of new antimycobacterial drugs. The regulation of host immune responses using immunoadjunctive agents may increase the efficacy of antimicrobial treatment against mycobacteriosis. In particular, the mild and long-term up-regulation of host immune reactions against mycobacterial pathogens using herbal medicines may be beneficial for such immunoadjunctive therapy. This review focuses on the current status regarding basic and clinical studies on protocols using herbal medicines, including medicinal plants, useful for the clinical treatment of intractable mycobacterial infections.
Collapse
Affiliation(s)
- Haruaki Tomioka
- Department of Basic Medical Science for Nursing, Department of Primary Education, Yasuda Women's University, Hiroshima 731-0153, Japan.
- Department of Microbiology and Immunology, Shimane University School of Medicine, Izumo 693-8501, Japan.
| | - Yutaka Tatano
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Otawara 324-8501, Japan.
| | - Toshiaki Shimizu
- Department of Nutritional Sciences, Yasuda Women's University, Hiroshima 731-0153, Japan.
| | - Chiaki Sano
- Department of Community Medicine Management, Shimane University School of Medicine, Izumo 693-8501, Japan.
| |
Collapse
|
26
|
Du X, Qian J, Wang Y, Zhang M, Chu Y, Li Y. Identification and immunological evaluation of novel TLR2 agonists through structure optimization of Pam 3CSK 4. Bioorg Med Chem 2019; 27:2784-2800. [PMID: 31101493 DOI: 10.1016/j.bmc.2019.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/26/2019] [Accepted: 05/04/2019] [Indexed: 12/22/2022]
Abstract
Toll-like receptor 2 (TLR2) is a bridge between innate immunity and adaptive immunity. TLR2 agonists have been exploited as potential vaccine adjuvants and antitumor agents. However, no TLR2 agonists have been approved by FDA up to now. To discover drug-like TLR2 selective agonists, a novel series of Pam3CSK4 derivatives were designed based on the crystal structure of hTLR2-hTLR1-Pam3CSK4 complex, synthesized and evaluated for their immune-stimulatory activities. Among them, 35c was identified as a murine-specific TLR2 agonist, while 35f was a human-specific TLR2 agonist. Besides, 35d (human and murine TLR2 agonist) showed TLR2 agonistic activity comparable to Pam3CSK4, which included: elevated IL-6 expression level (EC50 = 83.08 ± 5.94 nM), up-regulated TNF-α and IL-6 mRNA expression and promoted maturation of DCs through activating the NF-κB signaling pathway. TLRs antibodies test showed that 35a and 35d were TLR2/1 agonists, while 35f was a TLR2/6 agonist.
Collapse
Affiliation(s)
- Xinming Du
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiawen Qian
- Department of Immunology, School of Basic Medical Sciences and Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yujie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mingming Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences and Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Yingxia Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
27
|
Wu H, Wu Y, Ren L, Zhai W, Jiang Y, Guo S, Tao D, Su C, Chen Z, Jiang H. Effects of triptolide on bone marrow-derived mesenchymal stem cells from patients with multiple myeloma. Exp Ther Med 2019; 17:3291-3298. [PMID: 30988704 DOI: 10.3892/etm.2019.7373] [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: 11/25/2017] [Accepted: 08/17/2018] [Indexed: 12/16/2022] Open
Abstract
Triptolide (TPL), an extract of the Chinese herb Tripterygium wilfordii Hook F, is a potent anti-inflammatory agent that further possesses anticancer activity. Its antiproliferative effects are well established. Only few studies have focused on TPL as a potential treatment in multiple myeloma (MM). In the current study, bone marrow-derived mesenchymal stem cells (BMMSCs) from patients with MM were isolated and treated with TPL at varying concentrations. Thalidomide is currently used as a positive control drug in the treatment of MM. Cell Counting kit-8 assays were performed to assess proliferation activity and flow cytometry with Annexin V-fluorescein/propidium iodide was used to detect cell apoptosis of TPL-treated BMMSCs. Reverse transcription-quantitative polymerase chain reaction assays were applied to measure interleukin (IL)-6, IL-1β and stem cell factor (SCF or Kit ligand) mRNA expression and western blot assays were performed to analyze transcription factor p65 (P65) expression in TPL-treated BMMSCs. ELISA was applied to measure vascular endothelial growth factor (VEGF) levels in the supernatant of the cultured and treated BMMSCs. TPL treatment significantly inhibited BMMSC proliferation compared with the untreated control (P<0.05). At 48 h following TPL treatment, a Cell Counting kit-8 study was performed and the IC50 value was determined at 101.55±2.45 ng/ml. Apoptotic rates were observed to increase with increasing concentrations of TPL (P<0.001), and IL-6, IL-1β and SCF mRNA expression was significantly decreased with increasing TPL (P<0.001). P65 expression following TPL treatment was significantly decreased compared with the untreated control (P<0.05). VEGF levels were significantly reduced in the presence of increasing amounts of TPL (P<0.05). These findings suggest that TPL inhibited BMMSC growth and improved the bone marrow hematopoietic microenvironment by decreasing IL-6, IL-1β and SCF mRNA expression, subsequently inhibiting the proliferation of MM cells. Therefore, TPL may be used in the future to treat patients with MM.
Collapse
Affiliation(s)
- Haiying Wu
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Yuanting Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Li Ren
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Wo Zhai
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Yuxia Jiang
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Shuping Guo
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Diehong Tao
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Chuanyong Su
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Zhilu Chen
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Huifang Jiang
- Department of Hematology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
28
|
NFκB- and MAP-Kinase Signaling Contribute to the Activation of Murine Myeloid Dendritic Cells by a Flagellin A:Allergen Fusion Protein. Cells 2019; 8:cells8040355. [PMID: 30991709 PMCID: PMC6523117 DOI: 10.3390/cells8040355] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/26/2019] [Accepted: 04/12/2019] [Indexed: 01/22/2023] Open
Abstract
Fusion proteins incorporating the TLR5-ligand flagellin are currently undergoing clinical trials as vaccine candidates for many diseases. We recently reported a flagellin:allergen fusion protein containing the TLR5-ligand flagellin A (FlaA) from Listeria monocytogenes and the major birch pollen allergen Bet v 1 (rFlaA:Betv1) to prevent allergic sensitization in an experimental mouse model. This study analyzes the signaling pathways contributing to rFlaA:Betv1-mediated pro- and anti-inflammatory cytokine secretion and cell metabolism in myeloid dendritic cells (mDCs) in vitro. The influence of mammalian target of rapamycin (mTOR)-, NFκB-, and MAP kinase (MAPK)-signaling on cytokine secretion and metabolic activity of bone marrow (BM)-derived mDCs stimulated with rFlaA:Betv1 were investigated by pre-treatment with either mTOR- (rapamycin), NFκB- (dexamethason, BMS-345541, TPCA-1, triptolide, or BAY-11) or MAPK- (SP600125, U0126, or SB202190) inhibitors, respectively. rFlaA:Betv1-mediated IL-10 secretion as well as activation of mDC metabolism, rather than pro-inflammatory cytokine secretion, were inhibited by rapamycin. Inhibition of NFκB-signaling suppressed rFlaA:Betv1-induced IL-12, while inhibition of MAPK-signaling dose-dependently suppressed rFlaA:Betv1-induced IL-10 as well as pro-inflammatory IL-6 and TNF-α production. Notably, with the exception of a partial JNK-dependency, rFlaA:Betv1-mediated effects on mDC metabolism were mostly NFκB- and MAPK-independent. Therefore, MAPK-mediated activation of both NFκB- and mTOR-signaling likely is a key pathway for the production of pro- and anti-inflammatory cytokines by flagellin fusion protein vaccines.
Collapse
|
29
|
Zhao X, Tang X, Yan Q, Song H, Li Z, Wang D, Chen H, Sun L. Triptolide ameliorates lupus via the induction of miR-125a-5p mediating Treg upregulation. Int Immunopharmacol 2019; 71:14-21. [PMID: 30861393 DOI: 10.1016/j.intimp.2019.02.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/26/2022]
Abstract
Triptolide is a biologically active component of the Chinese antirheumatic herbal remedy Tripterygium wilfordii Hook F, which has been shown to be effective in treating murine lupus. However, its immunological mechanisms are poorly understood. Regulatory T cells (Treg) are pivotal for maintaining peripheral self-tolerance and controlling autoimmunity. This study was undertaken to examine the therapeutic effect of triptolide in lupus mice and the related molecular mechanisms. Our results showed that triptolide treatment ameliorated serum anti-dsDNA, proteinuria and renal histopathologic assessment in MRL/lpr mice, induced the miR-125a-5p expression and enhanced the proportion of Treg in vivo. In vitro, triptolide upregulated the proportion of Treg and the miR-125a-5p expression. Down-regulation of the miR-125a-5p expression reversed the effect of triptolide on Treg. In conclusion, triptolide could induce Treg and the miR-125a-5p expression in vivo and in vitro. Inhibiting the effect of miR-125a-5p could counteract the effect of triptolide on inducing Treg. The study has strong implications for the therapeutic applications of triptolide in systemic lupus erythematosus.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China; Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Qing Yan
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Hua Song
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Zutong Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Hongwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
30
|
Phan M, Watson MF, Alain T, Diallo JS. Oncolytic Viruses on Drugs: Achieving Higher Therapeutic Efficacy. ACS Infect Dis 2018; 4:1448-1467. [PMID: 30152676 DOI: 10.1021/acsinfecdis.8b00144] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Over the past 20 years there has been a dramatic expansion in the testing of oncolytic viruses (OVs) for the treatment of cancer. OVs are unique biotherapeutics that induce multimodal responses toward tumors, from direct cytopathic effects on cancer cells, to tumor associated blood vessel disruption, and ultimately potent stimulation of anti-tumor immune activation. These agents are highly targeted and can be efficacious as cancer treatments resulting in some patients experiencing complete tumor regression and even cures from OV monotherapy. However, most patients have limited responses with viral replication in tumors often found to be modest and transient. To augment OV replication, increase bystander killing of cancer cells, and/or stimulate stronger targeted anti-cancer immune responses, drug combination approaches have taken center stage for translation to the clinic. Here we comprehensively review drugs that have been combined with OVs to increase therapeutic efficacy, examining the proposed mechanisms of action, and we discuss trends in pharmaco-viral immunotherapeutic approaches currently being investigated.
Collapse
Affiliation(s)
- Michael Phan
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Margaret F. Watson
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Children’s Hospital of Eastern Ontario Research Institute, 401 Smyth Road Research Building 2, Second Floor, Room 2119, Ottawa, Ontario K1H 8L1, Canada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Children’s Hospital of Eastern Ontario Research Institute, 401 Smyth Road Research Building 2, Second Floor, Room 2119, Ottawa, Ontario K1H 8L1, Canada
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
31
|
Shang SL, Cai GY, Duan SW, Li P, Li QG, Chen XM. Retrospective analysis of tacrolimus combined with Tripterygium wilfordii polyglycoside for treating idiopathic membranous nephropathy. BMC Nephrol 2018; 19:182. [PMID: 30021637 PMCID: PMC6052665 DOI: 10.1186/s12882-018-0967-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Idiopathic membranous nephropathy (IMN) is one of the most common adult nephrotic syndromes. Some patients with this disorder require immunosuppressive therapy. This retrospective case series was performed to assess the effects of tacrolimus (TAC) combined with Tripterygium wilfordii polyglycoside (TWG) in treating IMN. METHODS From January 2015 to August 2016, kidney-biopsy-proven IMN patients treated with TAC in the Chinese PLA General Hospital were screened. Data were retrieved from the patients' medical records. The first efficacy evaluation index was remission rate (complete remission and partial remission), and the secondary efficacy evaluation indices included relapse rate, proteinuria, serum albumin and estimated glomerular filtration rate (eGFR). Adverse events were also assessed. RESULTS The included patients' treatments were tacrolimus monotherapy (TAC group, n = 33), tacrolimus combined with methylprednisolone (MP) (TAC + MP group, n = 24) and tacrolimus combined with Tripterygium wilfordii polyglycoside (TAC + TWG group, n = 21). The remission rates of the TAC, TAC + MP, and TAC + TWG groups in the 10th month were 54.5, 62.5, and 85.7%, respectively (TAC + TWG group vs TAC group, P = 0.037, TAC + TWG group vs TAC + MP group, P = 0.125). Moreover, the complete remission rates of the TAC, TAC + MP, and TAC + TWG groups in the 10th month were 21.2, 20.8, and 57.1%, respectively (TAC + TWG group vs TAC group, P = 0.007, TAC + TWG group vs TAC + MP group, P = 0.012). Compared with the TAC group, the TAC + TWG group had a higher remission rate during these ten months (log-rank, P = 0.005). Compared with the TAC and TAC + MP groups, the TAC + TWG group had a higher complete remission rate (log-rank, P = 0.019 and log-rank, P = 0.005, respectively). CONCLUSION This retrospective study showed that TAC combined with TWG may be effective for treating IMN. Further randomized controlled trials (RCTs) are needed to assess the efficacy and safety of TAC combined with TWG.
Collapse
Affiliation(s)
- Shun-Lai Shang
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Guang-Yan Cai
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shu-Wei Duan
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Ping Li
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Qing-Gang Li
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Xiang-Mei Chen
- Department of Nephrology, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
32
|
Zhang L, Shi L, Soars SM, Kamps J, Yin H. Discovery of Novel Small-Molecule Inhibitors of NF-κB Signaling with Antiinflammatory and Anticancer Properties. J Med Chem 2018; 61:5881-5899. [PMID: 29870245 DOI: 10.1021/acs.jmedchem.7b01557] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Excessive NF-κB activation contributes to the pathogenesis of numerous diseases. Small-molecule inhibitors of NF-κB signaling have significant therapeutic potential especially in treating inflammatory diseases and cancers. In this study, we performed a cell-based high-throughput screening to discover novel agents capable of inhibiting NF-κB signaling. On the basis of two hit scaffolds from the screening, we synthesized 69 derivatives to optimize the potency for inhibition of NF-κB activation, leading to successful discovery of the most potent compound Z9j with over 170-fold enhancement of inhibitory activity. Preliminary mechanistic studies revealed that Z9j inhibited NF-κB signaling via suppression of Src/Syk, PI3K/Akt, and IKK/IκB pathways. This novel compound also demonstrated antiinflammatory and anticancer activities, warranting its further development as a potential multifunctional agent to treat inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Chemistry and Biochemistry and the BioFrontiers Institute , University of Colorado , Boulder , Colorado 80309 , United States
| | - Lei Shi
- Department of Chemistry and Biochemistry and the BioFrontiers Institute , University of Colorado , Boulder , Colorado 80309 , United States.,Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 210009 , P. R. China
| | - Shafer Myers Soars
- Department of Chemistry and Biochemistry and the BioFrontiers Institute , University of Colorado , Boulder , Colorado 80309 , United States
| | - Joshua Kamps
- Department of Chemistry and Biochemistry and the BioFrontiers Institute , University of Colorado , Boulder , Colorado 80309 , United States
| | - Hang Yin
- Department of Chemistry and Biochemistry and the BioFrontiers Institute , University of Colorado , Boulder , Colorado 80309 , United States
| |
Collapse
|
33
|
Liu H, Tang L, Li X, Li H. Triptolide inhibits vascular endothelial growth factor-mediated angiogenesis in human breast cancer cells. Exp Ther Med 2018; 16:830-836. [PMID: 30116337 PMCID: PMC6090217 DOI: 10.3892/etm.2018.6200] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/15/2017] [Indexed: 12/12/2022] Open
Abstract
Triptolide has been demonstrated to induce tumor cell apoptosis. However, the role of triptolide in breast cancer angiogenesis remains unclear. The present study aimed to investigate the function of triptolide in breast cancer and the molecular mechanisms underlying this. The results revealed that triptolide could significantly decrease the expression of vascular endothelial growth factor A (VEGFA) in Hs578T and MDAMB231 breast cancer cells. Furthermore, human umbilical vein endothelial cells were used to perform tube formation and bromodeoxyuridine incorporation assays, which demonstrated an antiangiogenic effect of triptolide. In addition, the effect of triptolide in vivo was examined in a xenograft mouse model, which determined that VEGFA, cluster of differentiation 31 and anti-proliferation marker protein Ki67 expression in tumor sections was decreased in the triptolide treatment group compared with the control group. Western bolt analysis was performed to investigate the phosphorylation of extracellular signal-related kinase (ERK)1/2 and RAC-α serine/threonine-protein kinase after triptolide treatment, and it's effect on hypoxia inducible factor (HIF)1-α expression. The results demonstrated that triptolide suppressed ERK1/2 activation and HIF1-α expression. Furthermore, overexpression of HIF1-α could partially abrogate the inhibitory effect of triptolide on VEGFA expression. These results suggest that triptolide inhibits breast cancer cell angiogenesis in vitro and in vivo through inhibiting the ERK1/2-HIF1-α-VEGFA axis.
Collapse
Affiliation(s)
- Huantao Liu
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lubing Tang
- Department of Breast Surgery, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, P.R. China
| | - Xiaoyan Li
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Huiying Li
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
34
|
Zhou YZ, Zhao LD, Chen H, Zhang Y, Wang DF, Huang LF, Lv QW, Liu B, Li Z, Wei W, Li H, Liao X, Liu H, Liu X, Jin H, Wang J, Fei YY, Wu QJ, Zhang W, Shi Q, Zheng WJ, Zhang FC, Tang FL, Lipsky PE, Zhang X. Comparison of the impact of Tripterygium wilfordii Hook F and Methotrexate treatment on radiological progression in active rheumatoid arthritis: 2-year follow up of a randomized, non-blinded, controlled study. Arthritis Res Ther 2018; 20:70. [PMID: 29636089 PMCID: PMC5894170 DOI: 10.1186/s13075-018-1563-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/12/2018] [Indexed: 01/19/2023] Open
Abstract
Background Tripterygium wilfordii Hook F (TwHF) alone or in combination with methotrexate (MTX) has been shown to be more effective than MTX monotherapy in controlling the manifestations in subjects with disease-modifying antirheumatic drug (DMARD)-naïve active rheumatoid arthritis (RA) over a 6-month period. The long-term impact of these therapies on disease activity and radiographic progression in RA has not been examined. Methods Patients with DMARD-naïve RA enrolled in the “Comparison of Tripterygium wilfordii Hook F with methotrexate in the Treatment of Active Rheumatoid Arthritis” (TRIFRA) study were randomly allocated into three arms with TwHF or MTX or the two in combination. Clinical indexes and radiographic data at baseline and year 2 was collected and compared using an intent-to-treat (ITT) and a per-protocol (PP) analysis. Two radiologists blinded to the treatment scored the images independently. Results Of 207 subjects 109 completed the 2-year follow up. The number of subjects withdrawing from the study and the number adhering to the initial regimens were similar among the three groups (p > = 0.05). In the ITT analysis, proportions of patients reaching American College of Rheumatology 50% (ACR50) response criteria were 46.4%, 58.0% and 50.7% in the MTX, TwHF and MTX + TwHF groups (TwHF vs MTX monotherapy, p = 0.004). Similar patterns were found in ACR20, ACR70, Clinical Disease Activity Index good responses, European League Against Rheumatism good response, remission rate and low disease activity rate at year 2. The results of the PP analysis agreed with those in the ITT analysis. The changes in total Sharp scores and joint erosion and joint space narrowing during the 2 years were associated with changes in disease activity measured by the 28-joint count Disease Activity Score and were comparable among the three groups (p > 0.05). Adverse events were similar in the three treatment groups. Conclusions During the 2-year therapy period, TwHF monotherapy was not inferior to MTX monotherapy in controlling disease activity and retarding radiological progression in patients with active RA. Trial registration This is a follow-up study. Original trial registration: ClinicalTrials.gov, NCT01613079. Registered on 4 June 2012.
Collapse
Affiliation(s)
- Yang-Zhong Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Li-Dan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Yan Zhang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan-Feng Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin-Fang Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China.,Department of Nephrology and Rheumatology, The First People's Hospital of ChenZhou, ChenZhou, Hunan, China
| | - Qian-Wen Lv
- The Body Sculpture and Liposuction Center of Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100144, China
| | - Bin Liu
- Department of Rheumatology, The Affiliated Hospital of QingDao University Medical College, Qingdao, Shandong, China
| | - Zhenbin Li
- Department of Rheumatology, The Bethune International Heping Hospital of Hebei, Shijiazhuang, Hebei, China
| | - Wei Wei
- Department of Rheumatology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Hongbin Li
- Department of Rheumatology, Affiliated Hospital of Inner Mongolia Medical College, Hohhot, Inner Mongolia, China
| | - Xiangping Liao
- Department of Nephrology and Rheumatology, The First People's Hospital of ChenZhou, ChenZhou, Hunan, China
| | - Hui Liu
- Department of Rheumatology, Beijing Dongfang Hospital, Beijing, China
| | - Xiumei Liu
- Department of Rheumatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongtao Jin
- Department of Rheumatology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Junxiang Wang
- Department of Rheumatology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yun-Yun Fei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Qing-Jun Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Qun Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Wen-Jie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Feng-Chun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Fu-Lin Tang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Peter E Lipsky
- Formerly National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA. .,AMPEL BioSolutions, Charlottesville, VA, 22901, USA.
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
35
|
Hamdoun S, Efferth T. Ginkgolic acids inhibit migration in breast cancer cells by inhibition of NEMO sumoylation and NF-κB activity. Oncotarget 2018; 8:35103-35115. [PMID: 28402272 PMCID: PMC5471038 DOI: 10.18632/oncotarget.16626] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/03/2017] [Indexed: 11/25/2022] Open
Abstract
Ginkgolic acids (GA), a group of alkyl phenols found in crude extracts of Ginkgo biloba leaves, are known to have anticancer activity, but their mode of action is not well understood. Our aim in this study was to investigate the anti-migratory activity of seven GA against breast cancer cells and to determine the molecular mechanism behind this activity. All seven GA and their mixture inhibited wound healing in MCF-7 and MDA-MB 231 breast cancer cells. None of the compounds nor the mixture showed cytotoxicity towards the two cell lines, if tested by the resazurin assay. C13:0 inhibited NF-κB activity in the HEK Blue Null 1 reporter cell line. Furthermore, C13:0 inhibited degradation of nuclear factor of κ-light polypeptide gene enhancer in B-cells inhibitor α (IκBα). Sumoylation assay revealed that GA inhibited sumoylation of NF-κB essential modulator (NEMO). Molecular docking on SUMO-activating enzyme E1 showed that the seven GA bound to the active adenylation site with high calculated affinities ranging from -10.28 to -12.27 kcal/mol. Quantitative RT-PCR using C15:0, C13:0 and the mixture showed a significant down-regulation of urokinase plasminogen activator (uPA), plasminogen activator inhibitor-1 (PAI-1), C-X-C chemokine receptor type 4 (CXCR4) and matrix metalloproteinase 9 (MMP-9). We conclude that GA revealed considerable anti-migratory activity at non-cytotoxic concentrations, indicating anti-metastatic activity with low toxicity. This effect can be explained by the inhibition of NEMO sumoylation leading to inhibition of IκBα degradation and consequently a reduction of NF-κB activity, leading to the down-regulation of metastasis related genes including uPA, PAI-1, CXCR4, and MMP-9.
Collapse
Affiliation(s)
- Sami Hamdoun
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
36
|
Shubin NJ, Pham TN, Staudenmayer KL, Parent BA, Qiu Q, O'Keefe GE. A Potential Mechanism for Immune Suppression by Beta-Adrenergic Receptor Stimulation following Traumatic Injury. J Innate Immun 2018; 10:202-214. [PMID: 29455206 DOI: 10.1159/000486972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/17/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND β-Adrenergic agents suppress inflammation and may play an important role in posttraumatic infections. Mechanisms may include inhibition of MAP kinase signaling. We sought to determine whether MKP-1 contributed to catecholamine suppression of innate immunity and also wanted to know whether early catecholamine treatment after traumatic injury increases the risk of later nosocomial infection. METHODS We performed experiments using THP-1 cells and peripheral blood mononuclear cells from healthy individuals. We exposed cells to epinephrine and/or LPS and measured inflammatory gene transcription and MAP kinase activation. We inhibited MKP-1 activity to determine its role in catecholamine-induced immune suppression. Finally, we studied injured subjects to determine whether early catecholamine treatment was associated with nosocomial infection. RESULTS Epinephrine increases MKP-1 transcripts and protein and decreases LPS-induced p38 and JNK phosphorylation and TNF-α gene transcription. RNAi inhibition of MKP-1 at least partially restores LPS-induced TNF-α gene expression (p = 0.024). In the clinical cohort, subjects treated with β-adrenergic agents had an increased risk of ventilator-associated pneumonia (aOR = 1.9; 95% CI = 1.3-2.6) and bacteremia (aOR = 1.5; 95% CI = 1.1-2.3). CONCLUSIONS MKP-1 may have a role in catecholamine-induced suppression of innate immunity, and exogenous catecholamines might contribute to nosocomial infection risk.
Collapse
Affiliation(s)
- Nicholas J Shubin
- Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, Washington, USA
| | - Tam N Pham
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | | | - Brodie A Parent
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Qian Qiu
- Harborview Injury Prevention and Research Center, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Grant E O'Keefe
- Harborview Injury Prevention and Research Center, Department of Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
37
|
Das N, Dewan V, Grace PM, Gunn RJ, Tamura R, Tzarum N, Watkins LR, Wilson IA, Yin H. HMGB1 Activates Proinflammatory Signaling via TLR5 Leading to Allodynia. Cell Rep 2017; 17:1128-1140. [PMID: 27760316 DOI: 10.1016/j.celrep.2016.09.076] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/19/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022] Open
Abstract
Infectious and sterile inflammatory diseases are correlated with increased levels of high mobility group box 1 (HMGB1) in tissues and serum. Extracellular HMGB1 is known to activate Toll-like receptors (TLRs) 2 and 4 and RAGE (receptor for advanced glycation endproducts) in inflammatory conditions. Here, we find that TLR5 is also an HMGB1 receptor that was previously overlooked due to lack of functional expression in the cell lines usually used for studying TLR signaling. HMGB1 binding to TLR5 initiates the activation of NF-κB signaling pathway in a MyD88-dependent manner, resulting in proinflammatory cytokine production and pain enhancement in vivo. Biophysical and in vitro results highlight an essential role for the C-terminal tail region of HMGB1 in facilitating interactions with TLR5. These results suggest that HMGB1-modulated TLR5 signaling is responsible for pain hypersensitivity.
Collapse
Affiliation(s)
- Nabanita Das
- Department of Chemistry and Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Varun Dewan
- Department of Chemistry and Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Peter M Grace
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Robin J Gunn
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ryo Tamura
- Department of Chemistry and Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Netanel Tzarum
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hang Yin
- Department of Chemistry and Biochemistry and the BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
38
|
Tomioka H. Usefulness of Chinese Herbal Medicines as Host-Directed Therapeutics against Mycobacterial Infections: A Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1597-1611. [PMID: 29121801 DOI: 10.1142/s0192415x17500860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The high incidence of tuberculosis (TB) in developing countries, the resurgence of TB in industrialized countries, and the worldwide increase in the prevalence of Mycobacterium avium complex infections have prompted the quest for new antimycobacterial drugs. However, the development of such chemotherapeutics is currently making very slow progress. It therefore appears that devising improved administration protocols for clinical treatment against intractable mycobacteriosis using existing chemotherapeutics is more practical than awaiting the development of novel antimycobacterial drugs. The modulation of host immune responses using immunoadjunctive agents may increase the efficacy of antimicrobial treatment against mycobacteriosis. Particularly, the mild and long-term up-regulation of host immune reactions against mycobacterial pathogens using Chinese herbal medicines (CHMs) may be beneficial for immunoadjunctive therapy. This review focuses on the current status and future prospects regarding the development of CHMs that can be useful for the clinical control of intractable mycobacterial infections.
Collapse
Affiliation(s)
- Haruaki Tomioka
- 1 Department of Basic Medical Science for Nursing, Department of Primary Education, Yasuda Women's University, Hiroshima, Japan.,2 Shimane University School of Medicine, Izumo, Shimane, Japan
| |
Collapse
|
39
|
Sakamoto K, Okuwaki T, Ushikubo H, Mori A, Nakahara T, Ishii K. Activation inhibitors of nuclear factor kappa B protect neurons against the NMDA-induced damage in the rat retina. J Pharmacol Sci 2017; 135:S1347-8613(17)30162-7. [PMID: 29110956 DOI: 10.1016/j.jphs.2017.09.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/13/2017] [Indexed: 01/08/2023] Open
Abstract
We reported that high-mobility group Box-1 (HMGB1) was involved in excitoneurotoxicity in the retina. HMGB1 is known to activate nuclear factor kappa B (NF-κB). However, the role of NF-κB in excitotoxicity is still controversial. Here, we demonstrated that NF-κB activation induced by NMDA led to the retinal neurotoxicity. Male Sprague-Dawley rats were used, and NMDA (200 nmol/eye) and bovine HMGB1 (15 μg/eye) were intravitreally injected. Triptolide (500 pmol/eye), BAY 11-7082 (500 pmol/eye), and IMD-0354 (7.5 nmol/eye), NF-κB inhibitors, were co-injected with NMDA or HMGB1. Retinal sections were obtained seven days after intravitreal injection. Cell loss in the ganglion cell layer was observed in the HMGB1- and the NMDA-treated retina. All of the NF-κB inhibitors used in this study reduced the damage. BAY 11-7082 reduced the expression of phosphorylated NF-κB 12 h after NMDA injection, upregulation of GFAP immunoreactivity induced by NMDA 12 and 48 h after NMDA injection, and the number of TUNEL-positive cells 48 h after NMDA injection. The results suggest that NF-κB activation is one of the mechanisms of the retinal neuronal death that occurs 48 h after NMDA injection or later. Prevention of NF-kB activation is a candidate for the treatment of retinal neurodegeneration associated with excitotoxicity.
Collapse
Affiliation(s)
- Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan.
| | - Tatsuya Okuwaki
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Hiroko Ushikubo
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| | - Kunio Ishii
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo 108-8641, Japan
| |
Collapse
|
40
|
Qiu S, Lv D. Triptolide inhibits CD4 + memory T cell-mediated acute rejection and prolongs cardiac allograft survival in mice. Exp Ther Med 2017; 14:2817-2822. [PMID: 28912844 PMCID: PMC5585726 DOI: 10.3892/etm.2017.4867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 04/07/2017] [Indexed: 12/11/2022] Open
Abstract
There have been numerous investigations into the immunosuppressive effects of triptolide; however, its inhibitory effects on memory T cells remain to be elucidated. Using a cluster of differentiation (CD)4+ memory T-cell transfer model, the aim of the present study was to determine the inhibitory effects of triptolide on CD4+ memory T cell-mediated acute rejection and to determine the potential underlying mechanisms. At 4 weeks after skin transplantation, mouse cervical heart transplantation was performed following the transfer of CD4+ memory T cells. Mice were divided into two groups: A Control [normal saline, 30 ml/kg/day; intraperitoneal injection (ip)] and a triptolide group (triptolide, 3 mg/kg/day; ip). Graft survival, pathological examination and the corresponding International Society for Heart & Lung Transplantation (ISHLT) scores were assessed 5 days following heart transplantation, and levels of interleukin (IL)-2, interferon-γ (IFN-γ), IL-10 and transforming growth factor β1 (TGF-β1) in cardiac grafts and peripheral blood were assessed using reverse transcription-quantitative polymerase chain reaction and ELISA. The duration of cardiac graft survival in the triptolide group was significantly increased compared with the control group (14.3±0.4 vs. 5.3±0.2 days; P<0.001). Further pathological examinations revealed that the infiltration of inflammatory cells and myocardial damage in the cardiac grafts was notably reduced by triptolide, and the corresponding ISHLT scores in the triptolide group were significantly lower than those of the control group (grade 2.08±0.15 vs. 3.67±0.17; P<0.001). In addition, triptolide was able to significantly reduce IL-2 and IFN-γ secretion (P<0.01), significantly increase TGF-β1 secretion in the cardiac grafts and peripheral blood (P<0.01) and increase IL-10 secretion in the cardiac grafts. Therefore, the present study suggests that triptolide inhibits CD4+ memory T cell-mediated acute rejection and prolongs cardiac allograft survival in mice. This effect may be mediated by the inhibition of cytokine secretion by type 1 T helper cells and promotion of regulatory T cell proliferation.
Collapse
Affiliation(s)
- Shuiwei Qiu
- Department of Cardiothoracic Surgery, People's Hospital of Quzhou, Quzhou, Zhejiang 324000, P.R. China
| | - Dingliang Lv
- Department of Cardiothoracic Surgery, People's Hospital of Quzhou, Quzhou, Zhejiang 324000, P.R. China
| |
Collapse
|
41
|
Hu C, Wang Y, Liao Y, Wang J, Sun B. Metabolomic Analysis of Adipose Tissue in Rats Exposed to Triptolide. Chromatographia 2017. [DOI: 10.1007/s10337-017-3328-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
42
|
Jiang S, Yan TS, Han YC, Cui LQ, Xue XS, Zhang C. Hypervalent-Iodine-Mediated Formation of Epoxides from Carbon(sp2)–Carbon(sp3) Single Bonds. J Org Chem 2017; 82:11691-11702. [DOI: 10.1021/acs.joc.7b00883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Shan Jiang
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Tai-Shan Yan
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yong-Chao Han
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Li-Qian Cui
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiao-Song Xue
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chi Zhang
- State Key Laboratory of Elemento-Organic
Chemistry, Collaborative Innovation Center of Chemical Science and
Engineering, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
43
|
Sato T, Akimoto N, Takahashi A, Ito A. Triptolide suppresses ultraviolet B-enhanced sebum production by inhibiting the biosynthesis of triacylglycerol in hamster sebaceous glands in vivo and in vitro. Exp Ther Med 2017; 14:361-366. [PMID: 28672939 DOI: 10.3892/etm.2017.4461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/24/2017] [Indexed: 12/21/2022] Open
Abstract
Ultraviolet B (UVB) irradiation causes alterations in cutaneous barrier function, including excessive production of sebum in sebaceous glands, which is associated with the aggravation of acne. This study aimed to evaluate the inhibitory effects of triptolide, a diterpenoid triepoxide from Tripterygium wilfordii Hook F, on sebocytic lipogenesis in UVB-irradiated hamster skin in vivo and in vitro. Topical application of triptolide decreased the UVB-enhanced sebum accumulation in the sebaceous glands of hamster skin. The level of triacylglycerol (TG), a major sebum component, on the skin surface was reduced by triptolide treatment in UVB-irradiated hamsters, whereas there was no change in that of free-fatty acids and cholesterol, which are minor sebum components. UVB irradiation significantly enhanced TG production (P<0.01 in extracellular lipids, P<0.05 in intracellular lipids), and the activity of acyl coenzyme A/diacylglycerol acyltransferase (DGAT), a rate-limiting enzyme of TG synthesis, in differentiated hamster sebocytes (P<0.05 at 6 h and UVB of 0.62 kJ/m2, P<0.001 at 24 h and UVB 0.37 or 0.62 kJ/m2). Furthermore, triptolide significantly inhibited UVB-enhanced TG production (P<0.05 at 28 nM and P<0.01 at 56 and 112 nM triptolide) and DGAT activity (P<0.01 at 28 nM and P<0.001 at 56 and 112 nM triptolide) in differentiated hamster sebocytes. These results provide novel evidence that triptolide decreases UVB-enhanced sebum production by inhibiting DGAT-dependent TG biosynthesis in differentiated hamster sebocytes. These findings may be applicable to the prevention of acne aggravation.
Collapse
Affiliation(s)
- Takashi Sato
- Department of Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Noriko Akimoto
- Department of Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Aiko Takahashi
- Department of Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Akira Ito
- The Institute for Social Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
44
|
Kim SJ, Beak SM, Park SK. Supplementation with Triptolide Increases Resistance to Environmental Stressors and Lifespan in C. elegans. J Food Sci 2017; 82:1484-1490. [PMID: 28471052 DOI: 10.1111/1750-3841.13720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/16/2017] [Accepted: 03/27/2017] [Indexed: 11/29/2022]
Abstract
Triptolide is a major active compound found in Tripterygium wilfordii., also known as Thunder God Vine. Triptolide has been shown to have anti-inflammatory and anticancer activities. In this study, we examined the effect of dietary supplementation with triptolide on response to environmental stressors and lifespan in vivo using Caenorhabditis elegans as a model system. Treatment with 50 mg/L of triptolide in the growth media increased resistance to oxidative stress and reduced the generation of intracellular reactive oxygen species. We also observed a lifespan-extending activity for triptolide. Both mean and maximum lifespans were significantly increased by supplementation with triptolide. Response to other environmental stressors was modulated by triptolide. The survival after heat shock or UV irradiation was markedly increased in worms treated with triptolide. Unlike many lifespan-extending genetic or nutritional interventions, the longevity phenotype conferred by triptolide did not have the trade-off of a reduction in fertility or a delay in the gravid period. The expressions of hsp-16.2 and sod-3, known to positively correlate with a long lifespan, were significantly upregulated by supplementation with triptolide. These findings suggest that triptolide can exhibit antistress and lifespan-extending effects in vivo, possibly through its antioxidant activity and support the free radical theory of aging, which emphasizes the causative role of oxidative stress in aging.
Collapse
Affiliation(s)
- Sung-Jin Kim
- Dept. of Medical Biotechnology, College of Medical Sciences, Soonchunhyang Univ., Asan, Chungnam, Republic of Korea
| | - Sun-Mi Beak
- Dept. of Medical Biotechnology, College of Medical Sciences, Soonchunhyang Univ., Asan, Chungnam, Republic of Korea
| | - Sang-Kyu Park
- Dept. of Medical Biotechnology, College of Medical Sciences, Soonchunhyang Univ., Asan, Chungnam, Republic of Korea
| |
Collapse
|
45
|
Correspondence. World J Pediatr 2016; 12:376-379. [PMID: 27351577 DOI: 10.1007/s12519-016-0033-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
46
|
Wang Q, Meng J, Dong A, Yu JZ, Zhang GX, Ma CG. The Pharmacological Effects and Mechanism ofTripterygium wilfordiiHook F in Central Nervous System Autoimmunity. J Altern Complement Med 2016; 22:496-502. [DOI: 10.1089/acm.2016.0004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Qing Wang
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
| | - Jian Meng
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| | - Aiguo Dong
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
| | - Jie-zhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA
| | - Cun-Gen Ma
- 2011 Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Shanxi Province, People's Republic of China
- Institute of Brain Science, Shanxi Datong University, Datong, People's Republic of China
| |
Collapse
|
47
|
Bai S, Sun Y, Wu L, Wu Z, Fang M. [Tripotolide ameliorates inflammation and apoptosis induced by focal cerebral ischemia/reperfusion in rats]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2016; 45:493-500. [PMID: 28087909 PMCID: PMC10396981 DOI: 10.3785/j.issn.1008-9292.2016.09.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/07/2016] [Indexed: 11/15/2022]
Abstract
Objective: To investigate the effects of triptolide on inflammation and apoptosis induced by focal cerebral ischemia/reperfusion in rats. Methods: The rat model of focal cerebral ischemia/reperfusion injury was established according to Longa's method. A total of 80 SD rats were randomly divided into 5 groups:normal control, sham group, DMSO group, middle cerebral artery occlusion (MCAO) group, and MCAO with tripolide treatment group. TTC staining was used to examine the site and volume of cerebral infarction, and Longa score was employed for neurological disorders measurement. Number of astrocytes was measured by fluorescence staining, and neuronal apoptosis was determined by TUNEL staining. The expressions of inducible nitric oxide synthase(iNOS), cyclooxygenase 2(COX-2) and NF-κB proteins were detected by immunohistochemistry, and the expression of iNOS, COX-2 mRNA was detected by real-time PCR. Results: Compared with DMSO group and MCAO group, brain edema was improved (80.03±0.46)% (P<0.05), infarct volume was reduced (8.3±1.4)% (P<0.01), Longa score was decreased (1.38±0.20, P<0.05) in triptolide treatment group. Meanwhile triptolide also dramatically reduced the number of GFAP-positive astrocytes (P<0.05), alleviated protein expression of COX-2 (91.67±1.31), iNOS (95.24±5.07) and NF-κB (75.03±2.06) triggered by MCAO (all P<0.05), and induced a down-regulation of cell apoptosis as showed by TUNEL assay (64.15±3.52, P<0.05). Conclusion: Triptolide can reduce the cerebral infarction volume, attenuate brain edema and ameliorate the neurological deficits induced by cerebral ischemia-reperfusion injury rats, indicating that it might be used as a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Shi Bai
- School of Medicine, Taizhou University, Taizhou 318000, China; Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yayi Sun
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lijuan Wu
- School of Medicine, Taizhou University, Taizhou 318000, China
| | - Zhongmin Wu
- School of Medicine, Taizhou University, Taizhou 318000, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
48
|
Qiao Z, He M, He MU, Li W, Wang X, Wang Y, Kuai Q, Li C, Ren S, Yu Q. Synergistic antitumor activity of gemcitabine combined with triptolide in pancreatic cancer cells. Oncol Lett 2016; 11:3527-3533. [PMID: 27123146 DOI: 10.3892/ol.2016.4379] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/17/2016] [Indexed: 01/17/2023] Open
Abstract
Pancreatic cancer is a fatal human malignancy associated with an exceptionally poor prognosis. Novel therapeutic strategies are urgently required to treat this disease. In addition to immunosuppressive activity, triptolide possesses strong antitumor activity and synergistically enhances the antitumor activities of conventional chemotherapeutic drugs in preclinical models of pancreatic cancer. The present study investigated the antitumor effects of triptolide in pancreatic cancer cells, either in combination with gemcitabine, or alone. The pancreatic cancer BxPC-3 and PANC-1 cell lines were treated with triptolide, which resulted in time- and dose-dependent growth arrest. When incorporated into a sequential schedule, triptolide synergistically increased gemcitabine-induced cell growth inhibition and apoptosis, in addition to the cooperative regulation of B-cell lymphoma 2 family proteins and loss of mitochondrial membrane potential. Furthermore, triptolide enhanced gemcitabine-induced S phase arrest and DNA double-strand breaks, possibly through checkpoint kinase 1 suppression. The results of the present study suggest that triptolide has therapeutic potential for the treatment of pancreatic cancer, particularly when administered in combination with gemcitabine.
Collapse
Affiliation(s)
- Zhixin Qiao
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China; Medical Research Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Min He
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - M U He
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China; College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100022, P.R. China
| | - Weijing Li
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - Xuanlin Wang
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - Yanbing Wang
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China; College of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Qiyuan Kuai
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - Changlan Li
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China; College of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Suping Ren
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| | - Qun Yu
- Department of Blood Products and Substitutes, Beijing Institute of Transfusion Medicine, Beijing 100850, P.R. China
| |
Collapse
|
49
|
Glickman-Simon R, Steurich J. Acupuncture for Ischemic Stroke, Music for Anxiety in Mechanical Ventilation, Essential Fatty Acids for Depression, Mindfulness Meditation for Sleep in Older Adults, Tripterygium wilfordii for Rheumatoid Arthritis. Explore (NY) 2016; 12:71-5. [DOI: 10.1016/j.explore.2015.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Liu S, Li X, Li H, Liang Q, Chen J, Chen J. Comparison of tripterygium wilfordii multiglycosides and tacrolimus in the treatment of idiopathic membranous nephropathy: a prospective cohort study. BMC Nephrol 2015; 16:200. [PMID: 26637482 PMCID: PMC4669631 DOI: 10.1186/s12882-015-0199-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 11/28/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Idiopathic membranous nephropathy (IMN) is a major cause of nephrotic syndrome among adults. Considering the natural course of IMN, when to treat and with which immunosuppressive treatment need to be carefully considered in such patients. A combination of tripterygium wilfordii multiglycosides (TWG) and prednisone may be an effective option for treating patients with IMN. METHODS In this prospective cohort study, we enrolled patients with biopsy-proven IMN at our kidney centre. One cohort received TWG combined with prednisone, whereas another cohort received tacrolimus (TAC) combined with prednisone, for 36 weeks. The primary outcome was the remission rate, whereas the secondary outcomes included the time to remission, relapse rate, changes in serum albumin levels and daily urinary protein levels, estimated glomerular filtration rate, and adverse events. RESULTS A total of 53 patients with IMN met the criteria for enrollment, and all patients completed the therapy. At the end of the 36-week therapy, remission (either partial remission [PR] or complete remission [CR]) was observed in 20 patients (86.9 %) receiving TWG and in 27 patients (90.0 %) receiving TAC (p > 0.05), whereas CR was noted in 12 patients (52.2 %) receiving TWG and 14 patients (46.7 %) receiving TAC (p > 0.05). The probability of remission was similar for both the TWG and TAC groups (p > 0.05, by log-bank test). The mean time for achieving remission was 11.8 ± 12.5 weeks in the TWG group and 8.5 ± 9.1 weeks in the TAC group (p > 0.05). CONCLUSIONS The combination of TWG and predisone is an effective and safe therapy for IMN.
Collapse
Affiliation(s)
- Shanshan Liu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Xiayu Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Heng Li
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Qian Liang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Jun Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| |
Collapse
|