1
|
Zhang Y, Wu Q, Fu H, Pang J, Zhang Y, Zhou H, Zhuang L, Zhang X, Chen L, Yang Q. Kaempferol attenuates cyclosporine-induced renal tubular injury via inhibiting the ROS-ASK1-MAPK pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3001-3014. [PMID: 39316086 DOI: 10.1007/s00210-024-03409-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/21/2024] [Indexed: 09/25/2024]
Abstract
Cyclosporine (CSA) is a widely used immunosuppressive medication. CSA nephrotoxicity severely limits its application. Kaempferol (KPF), a naturally occurring phenolic compound, has a promising protective effect in reducing CSA-induced renal tubular injury, but the mechanism remains unknown. Our study aimed to determine the protective role of KPF against CSA-induced renal tubular injury. C57/B6 mice and the NRK-52E cell line were employed. CSA worsened renal function in mice, causing detachment and necrosis of tubular cells, leading to tubular vacuolation and renal interstitial fibrosis. CSA caused the detachment, rupture, and death of tubular cells in vitro, resulting in cell viability loss. KPF mitigated all these injurious alterations. KPF hindered CSA-induced ROS generation and protected renal tubular epithelial cells, similar to the antioxidant NAC. CSA lowered SOD activity and GSH levels while increasing MDA levels, and KPF ameliorated these changes. CSA caused phosphorylation of ASK1, JNK, and p38, similar to H2O2, whereas KPF significantly inhibited these changes. In conclusion, KPF reduces CSA-induced tubular epithelial cell injury via its antioxidant properties, inhibits the phosphorylation of ASK1, and inhibits the phosphorylation of p38 and JNK, implying that the synergistic use of KPF in CSA immunotherapy may be a promising option to reduce CSA-evoked renal injury.
Collapse
Affiliation(s)
- Yaowu Zhang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Qijing Wu
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Huali Fu
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Jieya Pang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Yiyuan Zhang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Hui Zhou
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Ling Zhuang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Xiaobo Zhang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China
| | - Lianhua Chen
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China.
| | - Qianqian Yang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu Province, China.
| |
Collapse
|
2
|
Li Y, Cui H, Li S, Li X, Guo H, Nandakumar KS, Li Z. Kaempferol modulates IFN-γ induced JAK-STAT signaling pathway and ameliorates imiquimod-induced psoriasis-like skin lesions. Int Immunopharmacol 2023; 114:109585. [PMID: 36527884 DOI: 10.1016/j.intimp.2022.109585] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Immune-mediated inflammation contributes to the development of psoriasis. However, long-term treatment with global immunosuppressive agents may cause a variety of side effects including recurrent infections. Kaempferol (KP), a natural flavonol, present in various plants is proposed to be useful for the treatment of psoriasis patients. Nevertheless, an explicit understanding of KP induced mechanisms is a prerequisite for its use in clinics. Therefore, we investigated the therapeutic effects and potential mode of action of KP using IFN-γ induced HaCaT cells and imiquimod-induced psoriasis-like skin lesions in mice. In this study, we found KP reduced intracellular ROS production, inhibited rhIFN-γ-induced IFN-γR1 expression, and up-regulated SOCS1 levels in HaCaT cells. In addition, KP inhibited rhIFN-γ-induced phosphorylation of JAK-STAT signaling molecules in HaCaT cells. Most importantly, KP alleviated imiquimod-induced psoriasis-like skin lesions in mice, histopathology and proportion of DCs in the skin. Besides, it reduced the population of γδT17 cells in the lymph nodes of the psoriatic mice and also decreased the gene expression of many proinflammatory cytokines, including interleukin IL-23, IL-17A, TNF-α, IL-6, and IL-1β in addition to down-regulation of the proinflammatory JAK-STAT signaling pathway. Thus, KP modulated IFN-γ induced JAK-STAT signaling pathway by inducing IFN-γR1 expression and up-regulating SOCS1 expression. In addition, KP also ameliorated imiquimod-induced psoriasis by reducing the dendritic cell numbers, and γδT17 cell population, along with down- modulation of the JAK-STAT pathway.
Collapse
Affiliation(s)
- Yanpeng Li
- School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Haodong Cui
- First Clinical School of Medicine, Inner Mongolia Medical University, 010110 Hohhot, China
| | - Shipeng Li
- School of Medicine, Kunming University of Science and Technology, 650093 Kunming, China
| | - Xingyan Li
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, 211199 Nanjing, China
| | - Hongtao Guo
- Nursing Department, Affiliated Hospital of Inner Mongolia Medical University, 010110 Hohhot, China
| | - Kutty Selva Nandakumar
- Department of Environmental and Biosciences, School of Business, Innovation and Sustainability, Halmstad University, 30118 Halmstad, Sweden; School of Pharmaceutical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Zhilei Li
- Clinical Pharmacy Division of Pharmacy Department, Southern University of Science and Technology Hospital, 518055 Shenzhen, China.
| |
Collapse
|
3
|
Tian H, Lin S, Wu J, Ma M, Yu J, Zeng Y, Liu Q, Chen L, Xu J. Kaempferol alleviates corneal transplantation rejection by inhibiting NLRP3 inflammasome activation and macrophage M1 polarization via promoting autophagy. Exp Eye Res 2021; 208:108627. [PMID: 34044014 DOI: 10.1016/j.exer.2021.108627] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/21/2021] [Accepted: 05/13/2021] [Indexed: 12/01/2022]
Abstract
Corneal transplantation rejection remains a major threat to the success rate of high-risk patients. Given the many side effects presented by traditional immunosuppressants, there is an urgency to clarify the mechanism of corneal transplantation rejection and to identify new therapeutic targets. Kaempferol is a natural flavonoid that has been proven in various studies to possess anti-inflammatory, antioxidant, anticancer, and neuroprotective properties. However, the effect of Ka on corneal transplantation remains largely unexplored. To address this, both at the in vivo and in vitro levels, we established a model of corneal allograft transplantation in Wistar rats and an LPS-induced inflammatory model using human THP-1-derived macrophages. In the transplantation experiments, we observed an enhancement of mRNA and protein level in the NLRP3/IL-1 β axis and in M1 macrophage polarization post-operation. In groups to which kaempferol intraperitoneal injections were administered, this response was effectively reduced. However, the effect of kaempferol was reversed after the application of autophagy inhibitors. Similarly, in the inflammatory model, we found that different concentrations of kaempferol reduced the LPS-induced M1 polarization and NLRP3 inflammasome activation. Moreover, we confirmed that kaempferol induced autophagy and that autophagy inhibitors reversed this effect in macrophages. In conclusion, we found that kaempferol can inhibit the activation of NLRP3 inflammasomes by inducing autophagy, thus inhibiting macrophage polarization, and ultimately alleviating corneal transplantation rejection. Thus, our study suggests that kaempferol is a potential therapeutic agent in the treatment of allograft rejection.
Collapse
Affiliation(s)
- Huiwen Tian
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shumei Lin
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jing Wu
- Department of Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Ming Ma
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jian Yu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuanping Zeng
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qi Liu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Linjiang Chen
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jing Xu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
4
|
Qiu F, Lu W, Ye S, Liu H, Zeng Q, Huang H, Liang CL, Chen Y, Zheng F, Zhang Q, Lu CJ, Dai Z. Berberine Promotes Induction of Immunological Tolerance to an Allograft via Downregulating Memory CD8 + T-Cells Through Altering the Gut Microbiota. Front Immunol 2021; 12:646831. [PMID: 33643325 PMCID: PMC7907598 DOI: 10.3389/fimmu.2021.646831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/26/2021] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence has linked the gut microbiota dysbiosis to transplant rejection while memory T-cells pose a threat to long-term transplant survival. However, it's unclear if the gut microbiome alters the formation and function of alloreactive memory T-cells. Here we studied the effects of berberine, a narrow-spectrum antibiotic that is barely absorbed when orally administered, on the gut microbiota, memory T-cells, and allograft survival. In this study, C57BL/6 mice transplanted with islets or a heart from BALB/c mice were treated orally with berberine. Allograft survival was observed, while spleen, and lymph node T-cells from recipient mice were analyzed using a flow cytometer. High-throughput sequencing and qPCR were performed to analyze the gut microbiota. CD8+ T-cells from recipients were cultured with the bacteria to determine potential T-cell memory cross-reactivity to a specific pathogen. We found that berberine suppressed islet allograft rejection, reduced effector CD8+CD44highCD62Llow and central memory CD8+CD44highCD62Lhigh T-cells (TCM), altered the gut microbiota composition and specifically lowered Bacillus cereus abundance. Further, berberine promoted long-term islet allograft survival induced by conventional costimulatory blockade and induced cardiac allograft tolerance as well. Re-colonization of B. cereus upregulated CD8+ TCM cells and reversed long-term islet allograft survival induced by berberine plus the conventional costimulatory blockade. Finally, alloantigen-experienced memory CD8+ T-cells from transplanted recipients rapidly responded to B. cereus in vitro. Thus, berberine prolonged allograft survival by repressing CD8+ TCM through regulating the gut microbiota. We have provided the first evidence that donor-specific memory T-cell generation is linked to a specific microbe and uncovered a novel mechanism underlying the therapeutic effects of berberine. This study may be implicated for suppressing human transplant rejection since berberine is already used in clinic to treat intestinal infections.
Collapse
Affiliation(s)
- Feifei Qiu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihui Lu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shulin Ye
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huazhen Liu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiaohuang Zeng
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiding Huang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chun-Ling Liang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuchao Chen
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fang Zheng
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qunfang Zhang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuan-Jian Lu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Ye S, Liu H, Chen Y, Qiu F, Liang CL, Zhang Q, Huang H, Wang S, Zhang ZD, Lu W, Dai Z. A Novel Immunosuppressant, Luteolin, Modulates Alloimmunity and Suppresses Murine Allograft Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:3436-3446. [PMID: 31732527 DOI: 10.4049/jimmunol.1900612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/09/2019] [Indexed: 02/07/2023]
Abstract
An allograft is rejected in the absence of any immunosuppressive treatment because of vigorous alloimmunity and thus requires extensive immunosuppression for its survival. Although there are many conventional immunosuppressants for clinical use, it is necessary to seek alternatives to existing drugs, especially in case of transplant patients with complicated conditions. Luteolin, a natural ingredient, exists in many plants. It exhibits multiple biological and pharmacological effects, including anti-inflammatory properties. In particular, luteolin has been shown to upregulate CD4+CD25+ regulatory T cells (Tregs) in the context of airway inflammation. However, it remains unknown whether luteolin regulates alloimmune responses. In this study, we demonstrated that luteolin significantly prolonged murine skin allograft survival, ameliorated cellular infiltration, and downregulated proinflammatory cytokine gene expression in skin allografts. Furthermore, luteolin increased the percentage of CD4+Foxp3+ Tregs while reducing frequency of mature dendritic cells and CD44highCD62Llow effector CD4+/CD8+ T cells posttransplantation. It also suppressed the proliferation of T cells and their production of cytokines IFN-γ and IL-17A in vitro while increasing IL-10 level in the supernatant. Moreover, luteolin promoted CD4+Foxp3+ Treg generation from CD4+CD25- T cells in vitro. Depleting Tregs largely, although not totally, reversed luteolin-mediated extension of allograft survival. More importantly, luteolin inhibited AKT/mTOR signaling in T cells. Thus, for the first time, to our knowledge, we found that luteolin is an emerging immunosuppressant as an mTOR inhibitor in allotransplantation. This finding could be important for the suppression of human allograft rejection, although it remains to be determined whether luteolin has an advantage over other conventional immunosuppressants in suppression of allograft rejection.
Collapse
Affiliation(s)
- Shulin Ye
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yuchao Chen
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Qunfang Zhang
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Haiding Huang
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Sumei Wang
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Zhong-De Zhang
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Weihui Lu
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| |
Collapse
|
6
|
Liu C, Liu H, Lu C, Deng J, Yan Y, Chen H, Wang Y, Liang C, Wei J, Han L, Dai Z. Kaempferol attenuates imiquimod-induced psoriatic skin inflammation in a mouse model. Clin Exp Immunol 2019; 198:403-415. [PMID: 31407330 PMCID: PMC6857081 DOI: 10.1111/cei.13363] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2019] [Indexed: 12/25/2022] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease that mainly affects the skin barrier. Treatment for psoriasis mainly includes conventional immunosuppressive drugs. However, long-term treatment with global immunosuppressive agents may cause a variety of side effects, including nephrotoxicity and infections. Kaempferol, a natural flavonol present in various plants, is known to possess potent anti-inflammatory, anti-oxidant and anti-cancerous properties. However, it is unknown whether kaempferol is also anti-psoriatic. Here we established an imiquimod (IMQ)-induced psoriatic mouse model to explore the potential therapeutic effects of kaempferol on psoriatic skin lesions and inflammation. In this study, we demonstrated that treatment with kaempferol protected mice from developing psoriasis-like skin lesions induced by topical administration of IMQ. Kaempferol reduced CD3+ T cell infiltration and gene expression of major proinflammatory cytokines, including interleukin (IL)-6, IL-17A and tumor necrosis factor (TNF)-α, in the psoriatic skin lesion. It also down-regulated proinflammatory nuclear factor kappa B (NF-κB) signaling in the skin. The therapeutic effects were associated with a significant increase in CD4+ forkhead box protein 3 (FoxP3)+ regulatory T cell (Treg ) frequency in the spleen and lymph nodes as well as FoxP3-positive staining in the skin lesion. Conversely, depletion of CD4+ CD25+ Tregs reversed the therapeutic effects of kaempferol on the skin lesion. Kaempferol also lowered the percentage of IL-17A+ CD4+ T cells in the spleen and lymph nodes of IMQ-induced psoriatic mice. Finally, kaempferol suppressed the proliferation of T cells in vitro and their mTOR signaling. Thus, our findings suggest that kaempferol may be a therapeutic drug for treating human psoriasis in the near future.
Collapse
Affiliation(s)
- C. Liu
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - H. Liu
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - C. Lu
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - J. Deng
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Y. Yan
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - H. Chen
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Y. Wang
- Department of Cancer BiologyBeckman Research Institute of the City of HopeDuarteCAUSA
| | - C.‐L. Liang
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - J. Wei
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - L. Han
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Z. Dai
- Section of Immunology and Joint Immunology Programthe Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
7
|
Zeng Q, Qiu F, Chen Y, Liu C, Liu H, Liang CL, Zhang Q, Dai Z. Shikonin Prolongs Allograft Survival via Induction of CD4 +FoxP3 + Regulatory T Cells. Front Immunol 2019; 10:652. [PMID: 30988670 PMCID: PMC6451963 DOI: 10.3389/fimmu.2019.00652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/11/2019] [Indexed: 01/16/2023] Open
Abstract
A transplanted organ is usually rejected without any major immunosuppressive treatment because of vigorous alloimmune responsiveness. However, continuous global immunosuppression may cause severe side effects, including nephrotoxicity, tumors, and infections. Therefore, it is necessary to seek novel immunosuppressive agents, especially natural ingredients that may provide sufficient efficacy in immunosuppression with minimal side effects. Shikonin is a bioactive naphthoquinone pigment, an ingredient originally extracted from the root of Lithospermum erythrorhizon. Previous studies have shown that shikonin regulates immunity and exerts anti-inflammatory effects. In particular, it can ameliorate arthritis in animal models. However, it is unclear whether shikonin inhibits alloimmunity or allograft rejection. In this study and for the first time, we demonstrated that shikonin significantly prolonged the survival of skin allografts in wild-type mice. Shikonin increased the frequencies of CD4+Foxp3+ regulatory T cells (Tregs) post-transplantation and induced CD4+Foxp3+ Tregs in vitro as well. Importantly, depleting the Tregs abrogated the extension of skin allograft survival induced by shikonin. It also decreased the frequencies of CD8+CD44highCD62Llow effector T cells and CD11c+CD80+/CD11c+CD86+ mature DCs after transplantation. Moreover, we found that shikonin inhibited the proliferation of T cells in vitro and suppressed their mTOR signaling. It also reduced the gene expression of pro-inflammatory cytokines, including IFNγ, IL-6, TNFα, and IL-17A, while increasing the gene expression of anti-inflammatory mediators IL-10, TGF-β1, and indoleamine-2, 3-dioxygenase (IDO) in skin allografts. Further, shikonin downregulated IDO protein expression in skin allografts and DCs in vitro. Taken together, shikonin inhibits allograft rejection via upregulating CD4+Foxp3+ Tregs. Thus, shikonin is a novel immunosuppressant that could be potentially used in clinical transplantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhenhua Dai
- Section of Immunology, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Li Y, Strick-Marchand H, Lim AI, Ren J, Masse-Ranson G, Dan Li, Jouvion G, Rogge L, Lucas S, Bin Li, Di Santo JP. Regulatory T cells control toxicity in a humanized model of IL-2 therapy. Nat Commun 2017; 8:1762. [PMID: 29176694 PMCID: PMC5701141 DOI: 10.1038/s41467-017-01570-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/29/2017] [Indexed: 01/10/2023] Open
Abstract
While patient selection and clinical management have reduced high-dose IL-2 (HDIL2) immunotherapy toxicities, the immune mechanisms that underlie HDIL2-induced morbidity remain unclear. Here we show that dose-dependent morbidity and mortality of IL-2 immunotherapy can be modeled in human immune system (HIS) mice. Depletion of human T cell subsets during the HDIL2 treatment reduces toxicity, pointing to the central function of T cells. Preferential expansion of effector T cells secondary to defective suppressive capacity of regulatory T (Treg) cells after HDIL2 therapy further underscores the importance of Treg in the maintenance of immune tolerance. IL-2 toxicity is induced by selective depletion or inhibition of Treg after LDIL2 therapy, and is ameliorated in HDIL2-treated HIS mice receiving the PIM-1 kinase inhibitor, Kaempferol. Modeling IL-2 pathophysiology in HIS mice offers a means to understand the functions of effector and regulatory T cells in immune-mediated toxicities associated with cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Li
- Institut Pasteur, Innate Immunity Unit, Immunology Department, 75724, Paris, France
- Inserm U1223, 75724, Paris, France
| | - Helene Strick-Marchand
- Institut Pasteur, Innate Immunity Unit, Immunology Department, 75724, Paris, France
- Inserm U1223, 75724, Paris, France
| | - Ai Ing Lim
- Institut Pasteur, Innate Immunity Unit, Immunology Department, 75724, Paris, France
- Inserm U1223, 75724, Paris, France
| | - Jiazi Ren
- Key Laboratory of Molecular Virology and Immunology, CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200025, Shanghai, China
| | - Guillemette Masse-Ranson
- Institut Pasteur, Innate Immunity Unit, Immunology Department, 75724, Paris, France
- Inserm U1223, 75724, Paris, France
| | - Dan Li
- Key Laboratory of Molecular Virology and Immunology, CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200025, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 200025, Shanghai, China
| | - Gregory Jouvion
- Institut Pasteur, Human Histopathology and Animal Models Unit, 75724, Paris, France
| | - Lars Rogge
- Institut Pasteur, Immunoregulation Unit, Immunology Department, 75724, Paris, France
| | - Sophie Lucas
- de Duve Institute, Université Catholique de Louvain, and WELBIO, B1200, Brussels, Belgium
| | - Bin Li
- Key Laboratory of Molecular Virology and Immunology, CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200025, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 200025, Shanghai, China
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Immunology Department, 75724, Paris, France.
- Inserm U1223, 75724, Paris, France.
| |
Collapse
|
9
|
Qiu F, Liu H, Liang CL, Nie GD, Dai Z. A New Immunosuppressive Molecule Emodin Induces both CD4 +FoxP3 + and CD8 +CD122 + Regulatory T Cells and Suppresses Murine Allograft Rejection. Front Immunol 2017; 8:1519. [PMID: 29167674 PMCID: PMC5682309 DOI: 10.3389/fimmu.2017.01519] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/26/2017] [Indexed: 12/23/2022] Open
Abstract
Due to vigorous alloimmunity, an allograft is usually rejected without any conventional immunosuppressive treatment. However, continuous global immunosuppression may cause severe side effects, including tumors and infections. Mounting evidence has shown that cyclosporine (CsA), a common immunosuppressant used in clinic, impedes allograft tolerance by dampening regulatory T cells (Tregs), although it inhibits allograft rejection at the same time. Therefore, it is necessary to seek an alternative immunosuppressive drug that spares Tregs with high efficiency in suppression but low toxicity. In this study, we investigated the capacity of emodin, an anthraquinone molecule originally extracted from certain natural plants, to prolong transplant survival in a mouse model and explored the cellular and molecular mechanisms underlying its action. We found that emodin significantly extended skin allograft survival and hindered CD3+ T cell infiltration in the allograft, accompanied by an increase in CD4+Foxp3+ and CD8+CD122+ Treg frequencies and numbers but a reduction in effector CD8+CD44highCD62Llow T cells in recipient mice. Emodin also inhibited effector CD8+ T cells proliferation in vivo. However, CD4+CD25+, but not CD8+CD122+, Tregs derived from emodin-treated recipients were more potent in suppression of allograft rejection than those isolated from control recipients, suggesting that emodin also enhances the suppressive function of CD4+CD25+ Tregs. Interestingly, depleting CD25+ Tregs largely reversed skin allograft survival prolonged by emodin while depleting CD122+ Tregs only partially abrogated the same allograft survival. Furthermore, we found that emodin hindered dendritic cell (DC) maturation and reduced alloantibody production posttransplantation. Finally, we demonstrated that emodin inhibited in vitro proliferation of T cells and blocked their mTOR signaling as well. Therefore, emodin may be a novel mTOR inhibitor that suppresses alloimmunity by inducing both CD4+FoxP3+ and CD8+CD122+ Tregs, suppressing alloantibody production, and hindering DC maturation. Thus, emodin is a newly emerging immunosuppressant and could be utilized in clinical transplantation in the future.
Collapse
Affiliation(s)
- Feifei Qiu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Huazhen Liu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chun-Ling Liang
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Golay D. Nie
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Zhenhua Dai
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
10
|
Kashyap D, Sharma A, Tuli HS, Sak K, Punia S, Mukherjee TK. Kaempferol - A dietary anticancer molecule with multiple mechanisms of action: Recent trends and advancements. J Funct Foods 2017; 30:203-219. [PMID: 32288791 PMCID: PMC7104980 DOI: 10.1016/j.jff.2017.01.022] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 01/01/2017] [Accepted: 01/09/2017] [Indexed: 02/08/2023] Open
Abstract
The consumption of diet-based naturally bioactive metabolites is preferred to synthetic material in order to avert health-associated disorders. Among the plant-derived polyphenols, kaempferol (KMF) is considered as a valuable functional food ingredient with a broad range of therapeutic applications such as anti-cancer, antioxidant and anti-inflammatory uses. KMF acts on a range of intracellular as well as extracellular targets involved in the cell signaling pathways that in turn are known to regulate the hallmarks of cancer growth progressions like apoptosis, cell cycle, invasion or metastasis, angiogenesis and inflammation. Importantly, the understanding of mechanisms of action of KMF-mediated therapeutic effects may help the scientific community to design novel strategies for the treatment of dreadful diseases. The current review summarizes the various types of molecular targets of KMF in cancer cells as well as other health-associated disorders. In addition, this review also highlights the absorption, metabolism and epidemiological findings.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India
| | - Ajay Sharma
- Department of Chemistry, Career Point University, Tikker - kharwarian, Hamirpur, Himachal Pradesh 176041, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala, Haryana, India
| | | | - Sandeep Punia
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala, Haryana, India
| | - Tapan K. Mukherjee
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala, Haryana, India
| |
Collapse
|
11
|
Lu C, Zeng YQ, Liu H, Xie Q, Xu S, Tu K, Dou C, Dai Z. Tanshinol suppresses cardiac allograft rejection in a murine model. J Heart Lung Transplant 2017; 36:227-236. [PMID: 27574736 DOI: 10.1016/j.healun.2016.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/23/2016] [Accepted: 07/24/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Achieving long-term cardiac allograft survival without continuous immunosuppression is highly desired in organ transplantation. Studies have shown that Salvia miltiorrhiza, an herb also known as danshen, improves microcirculation and is highly effective in treating coronary heart disease. Our objective is to determine whether tanshinol, an ingredient of danshen, improves cardiac allograft survival. METHODS Fully vascularized heterotopic heart transplantation was performed using BALB/c mice as donors and C57BL/6 mice as recipients, which were then treated with tanshinol and rapamycin. CD4+FoxP3+ regulatory T cells (Tregs) were quantified by flow analyses, whereas CCL22 was measured by real-time polymerase chain reaction and Western blotting. RESULTS We found that tanshinol significantly delayed cardiac allograft rejection. It promoted long-term allograft survival induced by rapamycin, a mammalian target-of-rapamycin (mTOR) inhibitor. Tanshinol increased CD4+FoxP3+ Treg numbers in cardiac allografts, but not spleens and lymph nodes, of recipient mice by enhancing chemokine CCL22 expression in cardiac allografts, especially cardiac dendritic cells. In contrast, rapamycin increased Treg numbers in both lymphoid organs and allografts, suggesting that it generally expands Tregs. Moreover, Tregs induced by rapamycin plus tanshinol were more potent in suppressing T-cell proliferation in vitro than those from untreated recipients. Neutralizing CCL22 hindered CD4+FoxP3+ Treg migration to cardiac allografts and reversed long-term allograft survival induced by tanshinol plus rapamycin. CONCLUSIONS Tanshinol suppresses cardiac allograft rejection by recruiting CD4+FoxP3+ Tregs to the graft, whereas rapamycin does so via expanding the Tregs. Thus, tanshinol cooperates with rapamycin to further extend cardiac allograft survival.
Collapse
Affiliation(s)
- Chuanjian Lu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Yu-Qun Zeng
- Section of Nephrology, the Second Affiliated Hospital, Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huazhen Liu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Qingfeng Xie
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Shengmei Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi׳an, Shaanxi, China
| | - Changwei Dou
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi׳an, Shaanxi, China; Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Liu X, Zeng YQ, Liang YZ, Zou C, Liu H, Qiu F, Liang CL, Jin XW, Su ZR, Dai Z. Medicinal herbs Fructus corni and Semen cuscutae suppress allograft rejection via distinct immune mechanisms. Oncotarget 2016; 7:35680-35691. [PMID: 27256977 PMCID: PMC5094954 DOI: 10.18632/oncotarget.9680] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/17/2016] [Indexed: 12/16/2022] Open
Abstract
Achieving long-term allograft survival without continuous global immunosuppression is highly desirable because constant immunosuppression causes severe side effects. Traditional Chinese medicine (TCM) has been utilized to treat numerous diseases for centuries. To seek novel immunosuppressive agents, we investigated several Chinese herbal formulas that have been shown to be effective in treating autoimmune diseases. C57BL/6 mice were transplanted with a skin graft from Balb/C donors and treated orally with the TCM. IL-12-expressing dendritic cells and CD4+FoxP3+ Tregs were quantified by flow cytometer while intragraft IL-12 gene expression was measured by real-time PCR. Here we identified a unique TCM, San Si formula, which contains three herbs: Fructus corni (FC), Fructus ligustri lucidi (FLL) and Semen cuscutae (SC). We found that either SC or FC, but not FLL, significantly prolonged skin allograft survival while SC plus FC or San Si formula further delayed allograft rejection compared to SC or FC alone. SC and FC, which did not contain cyclosporine and rapamycin, reduced graft-infiltrating T cells and suppressed their proliferation. Importantly, it was SC, but not FC, that induced CD4+FoxP3+ Tregs in recipients. Tregs induced by SC were also more potent in suppression. In contrast, FC repressed both intracellular IL-12 expression by intragraft DCs and IFNγ expression by graft-infiltrating T cells. Moreover, FC inhibited intragraft IL-12 gene expression. Depleting Tregs and providing exogenous IL-12 completely reversed allograft survival induced by SC plus FC. Thus, SC and FC synergistically suppress allograft rejection via distinct mechanisms.
Collapse
MESH Headings
- Allografts/cytology
- Allografts/drug effects
- Allografts/immunology
- Animals
- Cornus/chemistry
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Drug Synergism
- Drug Therapy, Combination/methods
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Forkhead Transcription Factors/metabolism
- Graft Rejection/drug therapy
- Graft Survival/immunology
- Humans
- Immunosuppression Therapy/methods
- Immunosuppressive Agents/therapeutic use
- Interleukin-12/metabolism
- Medicine, Chinese Traditional/methods
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Real-Time Polymerase Chain Reaction
- Skin Transplantation/adverse effects
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transplantation, Homologous/adverse effects
Collapse
Affiliation(s)
- Xusheng Liu
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | - Yu-Qun Zeng
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | - Yong-Zhuo Liang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | - Chuan Zou
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | - Huazhen Liu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Feifei Qiu
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Chun-Lin Liang
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Xiao-Wei Jin
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Zi-Ren Su
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
| | - Zhenhua Dai
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P.R. China
- Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
13
|
Meng C, Zhi X, Li C, Li C, Chen Z, Qiu X, Ding C, Ma L, Lu H, Chen D, Liu G, Cui D. Graphene Oxides Decorated with Carnosine as an Adjuvant To Modulate Innate Immune and Improve Adaptive Immunity in Vivo. ACS NANO 2016; 10:2203-2213. [PMID: 26766427 DOI: 10.1021/acsnano.5b06750] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Current studies have revealed the immune effects of graphene oxide (GO) and have utilized them as vaccine carriers and adjuvants. However, GO easily induces strong oxidative stress and inflammatory reaction at the site of injection. It is very necessary to develop an alternative adjuvant based on graphene oxide derivatives for improving immune responses and decreasing side effects. Carnosine (Car) is an outstanding and safe antioxidant. Herein, the feasibility and efficiency of ultrasmall graphene oxide decorated with carnosine as an alternative immune adjuvant were explored. OVA@GO-Car was prepared by simply mixing ovalbumin (OVA, a model antigen) with ultrasmall GO covalently modified with carnosine (GO-Car). We investigated the immunological properties of the GO-Car adjuvant in model mice. Results show that OVA@GO-Car can promote robust and durable OVA-specific antibody response, increase lymphocyte proliferation efficiency, and enhance CD4(+) T and CD8(+) T cell activation. The presence of Car in GO also probably contributes to enhancing the antigen-specific adaptive immune response through modulating the expression of some cytokines, including IL-6, CXCL1, CCL2, and CSF3. In addition, the safety of GO-Car as an adjuvant was evaluated comprehensively. No symptoms such as allergic response, inflammatory redness swelling, raised surface temperatures, physiological anomalies of blood, and remarkable weight changes were observed. Besides, after modification with carnosine, histological damages caused by GO-Car in lung, muscle, kidney, and spleen became weaken significantly. This study sufficiently suggest that GO-Car as a safe adjuvant can effectively enhance humoral and innate immune responses against antigens in vivo.
Collapse
Affiliation(s)
- Chunchun Meng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
| | - Xiao Zhi
- Institute of Nano Biomedicine and Engineering, Key Laboratory of Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Chao Li
- Institute of Nano Biomedicine and Engineering, Key Laboratory of Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Chuanfeng Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
| | - Zongyan Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , 48 Wenhui Road, Yangzhou 225009, P. R. China
| | - Lijun Ma
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine , 1111 Xianxia Road, Shanghai 200336, P. R. China
| | - Hongmin Lu
- Department of Oncology, Shanghai Renji Hospital, Shanghai Jiao Tong University School of Medicine , 160 Pujian Road, Shanghai 200127, P. R. China
| | - Di Chen
- Institute of Nano Biomedicine and Engineering, Key Laboratory of Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , 518 Ziyue Road, Shanghai 200241, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , 48 Wenhui Road, Yangzhou 225009, P. R. China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Key Laboratory of Thin Film and Microfabrication Technology of Ministry of Education, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|