1
|
Torunoglu ST, Zajda A, Tampio J, Markowicz-Piasecka M, Huttunen KM. Metformin derivatives - Researchers' friends or foes? Biochem Pharmacol 2023; 215:115743. [PMID: 37591450 DOI: 10.1016/j.bcp.2023.115743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
Metformin has been used for ages to treat diabetes mellitus due to its safety profile and low cost. However, metformin has variable pharmacokinetics in patients, and due to its poor oral absorption, the therapeutic doses are relatively high, causing unpleasant gastrointestinal adverse effects. Therefore, novel derivatives of metformin have been synthesized during the past decades. Particularly, after the mid-2000 s, when organic cation transporters were identified as the main metformin carriers, metformin derivatives have been under intensive investigation. Nevertheless, due to the biguanide structure, derivatives of metformin have been challenging to synthesize. Moreover, the mechanisms of metformin's action are not fully understood to date, and since it has multifunctional properties, the interests have switched to re-purposing for other diseases. Indeed, metformin derivatives have been demonstrated in many cases to be more effective than metformin itself and have the potential to be used in different diseases, including several types of cancers and neurodegenerative diseases. On the other hand, the pleiotropic nature of metformin and its derivatives can also create challenges. Not all properties are fit for all diseases. In this review, the history of the development of metformin-like compounds is summarized, and insights into their potential for future drug discovery are discussed.
Collapse
Affiliation(s)
- Sema Tuna Torunoglu
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Agnieszka Zajda
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | | | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
2
|
Jiang X, Lu Z, Zhang Q, Yu J, Han D, Liu J, Li P, Li F. Osthole: A potential AMPK agonist that inhibits NLRP3 inflammasome activation by regulating mitochondrial homeostasis for combating rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154640. [PMID: 36608498 DOI: 10.1016/j.phymed.2022.154640] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/16/2022] [Accepted: 12/31/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Osthole (OST), a characteristic coumarin compound in Angelicae pubescentis radix (APR), has shown potent efficacy in the treatment of rheumatoid arthritis (RA), but its specific targets and potential mechanism are limited. PURPOSE This study aimed to explore the potential targets and molecular mechanisms of OST against RA using computer-assisted techniques in combination with RA fibroblast-like synoviocytes (FLS) inflammation model and CIA rat model. METHODS Network pharmacology and molecular docking were applied to initially predict the potential targets of OST for the treatment of RA. Thereafter, TNFα was used to stimulate FLS to build an in vitro model of inflammation, combined with RNA-seq technology and molecular biology such as qPCR to investigate the anti-inflammatory effects and related mechanisms of OST. Finally, the anti-RA effect of OST was demonstrated by establishing a CIA rat model. RESULTS The network model results showed that the anti-RA effect of OST was mainly related to its anti-inflammatory effect, and AMPK was identified as a potential target for the potency of OST. In the TNFα-induced FLS cells, OST inhibited the secretion of FLS inflammatory factors, which was attributed to the ability of OST to activate AMPK to inhibit the activation of the NLRP3 inflammasome. Further, it was observed that the activation of AMPK by OST facilitated mitochondrial biogenesis, and corrected abnormal mitochondrial dynamics in FLS, which was favoured to the restoration of mitochondrial homeostasis, and further promoted the occurrence of apoptosis and the decrease of ROS in FLS. Consistent with in vivo studies, administration of OST significantly improved joint deformity and toe erythema, reduced arthritis index scores and inhibited synovial inflammation in CIA rats. CONCLUSION Our study proposed for the first time that AMPK, served as a potential target of OST, positively participated in the anti-RA therapeutic effect of OST. By regulating mitochondrial homeostasis and function, OST can effectively inhibit the activation of inflammasome and the secretion of inflammatory factors in vitro and in vivo, and finally achieve beneficial effects in the treatment of RA, which provides support and greater possibility to make further efforts on pharmacological research and clinical application of OST.
Collapse
Affiliation(s)
- Xiaoli Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zhuojian Lu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Qian Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jialin Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Dong Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jinhong Liu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China.
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China; College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
3
|
Chen CW, Hu S, Tsui KH, Hwang GS, Chen ST, Tang TK, Cheng HT, Yu JW, Wang HC, Juang HH, Wang PS, Wang SW. Anti-inflammatory Effects of Gossypol on Human Lymphocytic Jurkat Cells via Regulation of MAPK Signaling and Cell Cycle. Inflammation 2019; 41:2265-2274. [PMID: 30136021 DOI: 10.1007/s10753-018-0868-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gossypol, a natural polyphenolic compound extracted from cottonseed oil, has been reported to possess pharmacological properties via modulation cell cycle and immune signaling pathway. However, whether gossypol has anti-inflammatory effects against phytohemagglutinin (PHA)-induced cytokine secretion in T lymphocytes through similar mechanism remains unclear. Using the T lymphocytes Jurkat cell line, we found that PHA exposure caused dramatic increase in interleukin-2 (IL-2) mRNA expression as well as IL-2 secretion. All of these PHA-stimulated reactions were attenuated in a dose-dependent manner by being pretreated with gossypol. However, gossypol did not show any in vitro cytotoxic effect at doses of 5-20 μM. As a possible mechanism underlying gossypol action, such as pronounced suppression IL-2 release, robust decreased PHA-induced phosphorylation of p38 and c-Jun N-terminal kinase expressions was found with gossypol pretreatment, but not significant phosphorylation of extracellular signal-regulated kinase expression. Furthermore, gossypol could suppress the Jurkat cells' growth, which was associated with increased percentage of G1/S phase and decreased fraction of G2 phase in flow cytometry test. We conclude that gossypol exerts anti-inflammatory effects probably through partial attenuation of mitogen-activated protein kinase (phosphorylated JNK and p38) signaling and cell cycle arrest in Jurkat cells.
Collapse
Affiliation(s)
- Chien-Wei Chen
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Sindy Hu
- Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Division of Geriatric Urology, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan.,Bioinformation Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Guey-Shyang Hwang
- Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Szu-Tah Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tswen-Kei Tang
- Department of Nursing, National Quemoy University, Kinmen County, Taiwan
| | - Hao-Tsai Cheng
- Division of Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ju-Wen Yu
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsiao-Chiu Wang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Horng-Heng Juang
- Bioinformation Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan.,Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Paulus S Wang
- Medical Center of Aging Research, China Medical University Hospital, Taichung, Taiwan, Republic of China. .,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan. .,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China.
| | - Shyi-Wu Wang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China. .,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
4
|
Andjelković A, Mordas A, Bruinsma L, Ketola A, Cannino G, Giordano L, Dhandapani PK, Szibor M, Dufour E, Jacobs HT. Expression of the Alternative Oxidase Influences Jun N-Terminal Kinase Signaling and Cell Migration. Mol Cell Biol 2018; 38:e00110-18. [PMID: 30224521 PMCID: PMC6275184 DOI: 10.1128/mcb.00110-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/11/2018] [Accepted: 09/11/2018] [Indexed: 12/25/2022] Open
Abstract
Downregulation of Jun N-terminal kinase (JNK) signaling inhibits cell migration in diverse model systems. In Drosophila pupal development, attenuated JNK signaling in the thoracic dorsal epithelium leads to defective midline closure, resulting in cleft thorax. Here we report that concomitant expression of the Ciona intestinalis alternative oxidase (AOX) was able to compensate for JNK pathway downregulation, substantially correcting the cleft thorax phenotype. AOX expression also promoted wound-healing behavior and single-cell migration in immortalized mouse embryonic fibroblasts (iMEFs), counteracting the effect of JNK pathway inhibition. However, AOX was not able to rescue developmental phenotypes resulting from knockdown of the AP-1 transcription factor, the canonical target of JNK, nor its targets and had no effect on AP-1-dependent transcription. The migration of AOX-expressing iMEFs in the wound-healing assay was differentially stimulated by antimycin A, which redirects respiratory electron flow through AOX, altering the balance between mitochondrial ATP and heat production. Since other treatments affecting mitochondrial ATP did not stimulate wound healing, we propose increased mitochondrial heat production as the most likely primary mechanism of action of AOX in promoting cell migration in these various contexts.
Collapse
Affiliation(s)
- Ana Andjelković
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Amelia Mordas
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Lyon Bruinsma
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Annika Ketola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Giuseppe Cannino
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Luca Giordano
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Praveen K Dhandapani
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Marten Szibor
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eric Dufour
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Lim WM, Ito Y, Sakata-Sogawa K, Tokunaga M. CLIP-170 is essential for MTOC repositioning during T cell activation by regulating dynein localisation on the cell surface. Sci Rep 2018; 8:17447. [PMID: 30487641 PMCID: PMC6261991 DOI: 10.1038/s41598-018-35593-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 02/02/2023] Open
Abstract
The microtubule-organizing centre (MTOC) is repositioned to the centre of the contacted cell surface, the immunological synapse, during T cell activation. However, our understanding of its molecular mechanism remains limited. Here, we found that the microtubule plus-end tracking cytoplasmic linker protein 170 (CLIP-170) plays a novel role in MTOC repositioning using fluorescence imaging. Inhibition of CLIP-170 phosphorylation impaired both MTOC repositioning and interleukin-2 (IL-2) expression. T cell stimulation induced some fraction of dynein to colocalise with CLIP-170 and undergo plus-end tracking. Concurrently, it increased dynein in minus-end-directed movement. It also increased dynein relocation to the centre of the contact surface. Dynein not colocalised with CLIP-170 showed both an immobile state and minus-end-directed movement at a velocity in good agreement with the velocity of MTOC repositioning, which suggests that dynein at the immunological synapse may pull the microtubules and the MTOC. Although CLIP-170 is phosphorylated by AMP-activated protein kinase (AMPK) irrespective of stimulation, phosphorylated CLIP-170 is essential for dynein recruitment to plus-end tracking and for dynein relocation. This indicates that dynein relocation results from coexistence of plus-end- and minus-end-directed translocation. In conclusion, CLIP-170 plays an indispensable role in MTOC repositioning and full activation of T cells by regulating dynein localisation.
Collapse
Affiliation(s)
- Wei Ming Lim
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, Midori, Yokohama, Kanagawa, 226-8501, Japan
| | - Yuma Ito
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, Midori, Yokohama, Kanagawa, 226-8501, Japan
| | - Kumiko Sakata-Sogawa
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, Midori, Yokohama, Kanagawa, 226-8501, Japan.
| | - Makio Tokunaga
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, Midori, Yokohama, Kanagawa, 226-8501, Japan.
| |
Collapse
|
6
|
Jo D, Park R, Kim H, Jang M, Lee EJ, Jang IS, Park J. AMP-activated protein kinase regulates the expression of human telomerase reverse transcriptase. PLoS One 2018; 13:e0207864. [PMID: 30475873 PMCID: PMC6257937 DOI: 10.1371/journal.pone.0207864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/07/2018] [Indexed: 12/25/2022] Open
Abstract
The expression of hTERT in tumor cells contributes to oncogenic transformation by promoting immortalization. For this reason, hTERT is one of the major targets for cancer therapy, and an efficient method to downregulate hTERT expression is required for treatment of hTERT-positive cancer. In this report, we demonstrated that inhibition of AMP-activated protein kinase (AMPK) downregulates the expression of hTERT. We screened cell signaling pathways in AMPK α1 knockout cells and found that AMPKα1 is required for activity of the hTERT promoter. AMPKα1 knockout cells showed decreased expression of hTERT mRNA and protein. We also demonstrated that compound C, a reversible AMPK inhibitor, suppressed the expression of hTERT. However, AMPK activators, including AICAR and metformin, did not increase the level of hTERT protein. Finally, we showed that tumor cells stably expressing hTERT are resistant to compound C treatment. These results indicate that AMPK activity is required for tumor progression.
Collapse
Affiliation(s)
- Daum Jo
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Rackhyun Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Hyunju Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Minsu Jang
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, Chung-Ang University School of Medicine, Seoul, Republic of Korea
| | - Ik-Soon Jang
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
- * E-mail:
| |
Collapse
|
7
|
Li QR, Ni WP, Lei NJ, Yang JY, Xuan XY, Liu PP, Gong GM, Yan F, Feng YS, Zhao R, Du Y. The overexpression of Fra1 disorders the inflammatory cytokine secretion by mTEC of myasthenia gravis thymus. Scand J Immunol 2018; 88:e12676. [PMID: 29807388 DOI: 10.1111/sji.12676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/22/2018] [Indexed: 12/01/2022]
Abstract
The thymus of a myasthenia gravis (MG) patient is often accompanied by and effected with follicular hyperplasia. Inflammatory cytokines in thymus induce the formation of germinal centres (GC). MG thymic inflammatory cytokines are predominantly secreted by stromal cells. Our previous studies revealed that the expression level of the Fra1 protein, which is a Fos member of the activator protein 1 transcription factors (AP-1), was higher in the MG thymus compared with that of the normal thymus. Based on that, we demonstrated that Fra1 was mainly expressed in medulla thymic epithelial cells (mTECs) and that the rate of Fra1 positive mTECs in the MG thymus was higher than normal. In vitro, we found that the expression of CCL-5, CCL-19 and CCL-21 could be regulated by Fra1 in mTEC and that IL-1β, IL-6, IL-8 and ICAM1 were downregulated in the Fra1 overexpression group and upregulated in the Fra1 knock-down group. Meanwhile, we detected that the expression levels of suppressor of cytokine signalling 3 (SOCS3) were significantly upregulated along with the overexpression of Fra1. Hence, we considered that the overexpression of Fra1 disrupted inflammatory cytokine secretion by mTEC in the MG thymus and that STAT3 and SOCS3 were strongly involved in this process.
Collapse
Affiliation(s)
- Q-R Li
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - W-P Ni
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - N-J Lei
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - J-Y Yang
- College of Veterinary Medicine, North West Agriculture and Forestry University, Zhengzhou, China
| | - X-Y Xuan
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - P-P Liu
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - G-M Gong
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - F Yan
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - Y-S Feng
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - R Zhao
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| | - Y Du
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Rao E, Zhang Y, Li Q, Hao J, Egilmez NK, Suttles J, Li B. AMPK-dependent and independent effects of AICAR and compound C on T-cell responses. Oncotarget 2017; 7:33783-95. [PMID: 27177226 PMCID: PMC5085118 DOI: 10.18632/oncotarget.9277] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/27/2016] [Indexed: 01/02/2023] Open
Abstract
As a master metabolic sensor, AMP-activated protein kinase (AMPK) is involved in different fundamental cellular processes. Regulation of AMPK activity either by agonists (e.g., AICAR) or by antagonists (e.g., Compound C) has been widely employed to study the physiological functions of AMPK. However, mounting evidence indicates AMPK-independent effects for these chemicals and how they regulate immune cell functions remains largely unknown. Herein, using T cells from AMPK conditional knockout mice and their wild type littermates, we demonstrate that AICAR and Compound C can, indeed, activate or inhibit AMPK activity in T cells, respectively. Specifically, AICAR inhibits, but Compound C promotes, Ca2+-induced T cell death in an AMPK-dependent manner. In contrast, our data also demonstrate that AICAR and Compound C inhibit T cell activation and cytokine production in an AMPK-independent manner. Moreover, we find that the AMPK-independent activity of AICAR and Compound Cis mediated via the mTOR signaling pathway in activated T cells. Our results not only reveal the critical role of AMPK in regulating T cell survival and function, but also demonstrate AMPK-dependent and independent rolesof AICAR/Compound C in regulating T cell responses, thus suggesting a context-dependent effect of these “AMPK regulators”.
Collapse
Affiliation(s)
- Enyu Rao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Yuwen Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Qiang Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong, University, Jinan, Shandong, China
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Jill Suttles
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
9
|
Lin S, Sun L, Lyu X, Ai X, Du D, Su N, Li H, Zhang L, Yu J, Yuan S. Lactate-activated macrophages induced aerobic glycolysis and epithelial-mesenchymal transition in breast cancer by regulation of CCL5-CCR5 axis: a positive metabolic feedback loop. Oncotarget 2017; 8:110426-110443. [PMID: 29299159 PMCID: PMC5746394 DOI: 10.18632/oncotarget.22786] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/16/2017] [Indexed: 12/29/2022] Open
Abstract
Aberrant energy metabolism is critical for cancer progression. Tumor-associated macrophages (TAMs) can stimulate tumor angiogenesis and enhance cancer metastasis; however, the metabolic interaction between cancer cells and macrophages characterized by lactate shuttles remains unclear. Here, we showed that lactate activated human macrophages to a TAM-like phenotype and stimulated the secretion of CCL5 by activation of Notch signaling in macrophages. Reciprocally, CCL5 increased cell migration, induced cancer cell EMT, and promoted aerobic glycolysis in breast cancer cells, suggesting a positive metabolic feedback loop in the co-culture system. Inhibition of CCR5, the cognate receptor of CCL5, or neutralization of CCL5, broke the metabolic loop and decreased cancer cell migration and EMT. Inhibition of aerobic glycolysis significantly reduced breast cancer cell EMT, indicated that aerobic glycolysis was necessary for the invasive phenotype of cancer cells. We further showed that TGF-β signaling regulated the expression of CCR5 in the co-culture system, and CCL5 induced glycolysis by mediation of AMPK signaling. The expression of CCL5-CCR5 axis was highly associated with macrophage infiltration, TGF-β and p-AMPK in clinical samples. CCL5-CCR5 axis promoted breast cancer metastasis in vivo. Our findings suggested a pivotal role of CCL5-CCR5 axis in the metabolic communication between cancer cells and macrophages.
Collapse
Affiliation(s)
- Sensen Lin
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Li Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaodan Lyu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiongfei Ai
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Danyu Du
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Nan Su
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Hongyang Li
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Luyong Zhang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jun Yu
- Department of Molecular Biology, Jiangsu Cancer Hospital, Nanjing 210009, PR China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
10
|
Voelkl J, Alesutan I, Primessnig U, Feger M, Mia S, Jungmann A, Castor T, Viereck R, Stöckigt F, Borst O, Gawaz M, Schrickel JW, Metzler B, Katus HA, Müller OJ, Pieske B, Heinzel FR, Lang F. AMP-activated protein kinase α1-sensitive activation of AP-1 in cardiomyocytes. J Mol Cell Cardiol 2016; 97:36-43. [PMID: 27106803 DOI: 10.1016/j.yjmcc.2016.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/13/2016] [Accepted: 04/18/2016] [Indexed: 01/12/2023]
Abstract
AMP-activated protein kinase (Ampk) regulates myocardial energy metabolism and plays a crucial role in the response to cell stress. In the failing heart, an isoform shift of the predominant Ampkα2 to the Ampkα1 was observed. The present study explored possible isoform specific effects of Ampkα1 in cardiomyocytes. To this end, experiments were performed in HL-1 cardiomyocytes, as well as in Ampkα1-deficient and corresponding wild-type mice and mice following AAV9-mediated cardiac overexpression of constitutively active Ampkα1. As a result, in HL-1 cardiomyocytes, overexpression of constitutively active Ampkα1 increased the phosphorylation of Pkcζ. Constitutively active Ampkα1 further increased AP-1-dependent transcriptional activity and mRNA expression of the AP-1 target genes c-Fos, Il6 and Ncx1, effects blunted by Pkcζ silencing. In HL-1 cardiomyocytes, angiotensin-II activated AP-1, an effect blunted by silencing of Ampkα1 and Pkcζ, but not of Ampkα2. In wild-type mice, angiotensin-II infusion increased cardiac Ampkα1 and cardiac Pkcζ protein levels, as well as c-Fos, Il6 and Ncx1 mRNA expression, effects blunted in Ampkα1-deficient mice. Pressure overload by transverse aortic constriction (TAC) similarly increased cardiac Ampkα1 and Pkcζ abundance as well as c-Fos, Il6 and Ncx1 mRNA expression, effects again blunted in Ampkα1-deficient mice. AAV9-mediated cardiac overexpression of constitutively active Ampkα1 increased Pkcζ protein abundance and the mRNA expression of c-Fos, Il6 and Ncx1 in cardiac tissue. In conclusion, Ampkα1 promotes myocardial AP-1 activation in a Pkcζ-dependent manner and thus contributes to cardiac stress signaling.
Collapse
Affiliation(s)
- Jakob Voelkl
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Ioana Alesutan
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Uwe Primessnig
- Department of Cardiology, Charité, Campus Virchow & German Centre for Cardiovascular Research (DZHK), Charite & Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Martina Feger
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Sobuj Mia
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Andreas Jungmann
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg, Germany, and DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Tatsiana Castor
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Robert Viereck
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Florian Stöckigt
- Department of Medicine - Cardiology, University Hospital Bonn, Sigmund-Freud-Str.25, 53127 Bonn, Germany
| | - Oliver Borst
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany
| | - Jan Wilko Schrickel
- Department of Medicine - Cardiology, University Hospital Bonn, Sigmund-Freud-Str.25, 53127 Bonn, Germany
| | - Bernhard Metzler
- Department of Medicine - Cardiology, Medical University Innsbruck, Anichstr.35, 6020 Innsbruck, Austria
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg, Germany, and DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg, Germany, and DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Burkert Pieske
- Department of Cardiology, Charité, Campus Virchow & German Centre for Cardiovascular Research (DZHK), Charite & Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Department of Cardiology, University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Frank R Heinzel
- Department of Cardiology, Charité, Campus Virchow & German Centre for Cardiovascular Research (DZHK), Charite & Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Florian Lang
- Department of Physiology & Cardiology and Cardiovascular Medicine, University of Tübingen, Gmelinstr.5/Otfried-Mueller-Str. 10, 72076, Tübingen, Germany.
| |
Collapse
|
11
|
Finley J. Reactivation of latently infected HIV-1 viral reservoirs and correction of aberrant alternative splicing in the LMNA gene via AMPK activation: Common mechanism of action linking HIV-1 latency and Hutchinson–Gilford progeria syndrome. Med Hypotheses 2015; 85:320-32. [DOI: 10.1016/j.mehy.2015.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 05/25/2015] [Accepted: 06/08/2015] [Indexed: 12/30/2022]
|
12
|
Extracellular signal-regulated kinase is a direct target of the anti-inflammatory compound amentoflavone derived from Torreya nucifera. Mediators Inflamm 2013; 2013:761506. [PMID: 23970815 PMCID: PMC3736407 DOI: 10.1155/2013/761506] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/24/2013] [Indexed: 01/02/2023] Open
Abstract
Amentoflavone is a biflavonoid compound with antioxidant, anticancer, antibacterial, antiviral, anti-inflammatory, and UV-blocking activities that can be isolated from Torreya nucifera, Biophytum sensitivum, and Selaginella tamariscina. In this study, the molecular mechanism underlying amentoflavone's anti-inflammatory activity was investigated. Amentoflavone dose dependently suppressed the production of nitric oxide (NO) and prostaglandin E2 (PGE2) in RAW264.7 cells stimulated with the TLR4 ligand lipopolysaccharide (LPS; derived from Gram-negative bacteria). Amentoflavone suppressed the nuclear translocation of c-Fos, a subunit of activator protein (AP)-1, at 60 min after LPS stimulation and inhibited the activity of purified and immunoprecipitated extracellular signal-regulated kinase (ERK), which mediates c-Fos translocation. In agreement with these results, amentoflavone also suppressed the formation of a molecular complex including ERK and c-Fos. Therefore, our data strongly suggest that amentoflavone's immunopharmacological activities are due to its direct effect on ERK.
Collapse
|
13
|
Kang KY, Kim YK, Yi H, Kim J, Jung HR, Kim IJ, Cho JH, Park SH, Kim HY, Ju JH. Metformin downregulates Th17 cells differentiation and attenuates murine autoimmune arthritis. Int Immunopharmacol 2013; 16:85-92. [PMID: 23557965 DOI: 10.1016/j.intimp.2013.03.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/13/2013] [Accepted: 03/21/2013] [Indexed: 12/14/2022]
Abstract
INTRODUCTION This study was undertaken to determine whether metformin has anti-inflammatory effects in the collagen antibody-induced arthritis (CAIA) murine model. The effect of metformin on Th17 cell differentiation was also investigated. METHODS CAIA mice were treated with 100 and 150 mg/kg i.p. metformin (low- and high-dose groups, respectively). Arthritis activity and histological joint destruction were studied. Flow cytometry was used to (i) determine RORγt-expressing CD4+ percentages in draining axillary lymph nodes (ALNs) from metformin-treated and untreated mice with CAIA, (ii) determine Th17 percentages in splenic CD4+ T cells cultured ex vivo for 3 days in Th17-differentiation-inducing conditions, and (iii) determine the percentages of RORγt+CD4+ T cells when normal splenic T cells from DBA/1 mice were cultured in Th17-differentiation-inducing conditions together with various metformin doses. Western blot analysis was used to assess the intracellular signaling of the metformin-treated splenocytes. RESULTS Metformin attenuated both arthritis scores and bone destruction in CAIA mice, decreased the serum levels of the pro-inflammatory cytokines, TNF-α and IL-1, and reduced the number of RORγt+CD4+ T cells in the ALNs. Splenocytes from metformin-treated CAIA mice differentiated less readily into Th17 cells upon ex vivo stimulation. Metformin treatment of normal cells cultured in Th17-differentiation-inducing conditions decreased the number of RORγt-expressing CD4+ cells in a dose-dependent manner and downregulated STAT3 phosphorylation via the AMPK pathway. CONCLUSIONS Metformin had an anti-inflammatory effect on murine autoimmune arthritis due to the inhibition of Th17 cell differentiation. Metformin may have a possible therapeutic value for treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Kwi Young Kang
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lee JY, Choi AY, Oh YT, Choe W, Yeo EJ, Ha J, Kang I. AMP-activated protein kinase mediates T cell activation-induced expression of FasL and COX-2 via protein kinase C theta-dependent pathway in human Jurkat T leukemia cells. Cell Signal 2012; 24:1195-207. [PMID: 22330070 DOI: 10.1016/j.cellsig.2012.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/27/2012] [Accepted: 01/27/2012] [Indexed: 12/17/2022]
Abstract
AMP-activated protein kinase (AMPK), an important regulator of energy homeostasis, is known to be activated during T cell activation. T cell activation by T cell receptor (TCR) engagement or its pharmacological mimics, PMA plus ionomycin (PMA/Io), induces immunomodulatory FasL and cyclooxygenase-2 (COX-2) expression. In this study, we examined the role and mechanisms of AMPK in PMA/Io-induced expression of FasL and COX-2 in Jurkat T human leukemic cells. Inhibition of AMPK by a pharmacological agent, compound C, or AMPKα1 siRNA suppressed expression of FasL and COX-2 mRNAs and proteins in PMA/Io-activated Jurkat cells. It also reduced secretion of FasL protein and prostaglandin E2, a main product of COX-2, in Jurkat cells and peripheral blood lymphocytes activated with PMA/Io or monoclonal anti-CD3 plus anti-CD28. Consistently, inhibition of AMPK blocked promoter activities of FasL and COX-2 in activated Jurkat cells. As protein kinase C theta (PKCθ) is a central molecule for TCR signaling, we examined any possible cross-talk between AMPK and PKCθ in activated T cells. Of particular importance, we found that inhibition of AMPK blocked phosphorylation and activation of PKCθ, suggesting that AMPK is an upstream kinase of PKCθ. Moreover, we showed that AMPK was directly associated with PKCθ and phosphorylated Thr538 of PKCθ in PMA/Io-stimulated Jurkat cells. We also showed that inhibition of PKCθ by rottlerin or dominant negative PKCθ reduced AMPK-mediated transcriptional activation of NF-AT and AP-1 in activated Jurkat cells. Taken together, these results suggest that AMPK regulates expression of FasL and COX-2 via the PKCθ and NF-AT and AP-1 pathways in activated Jurkat cells.
Collapse
Affiliation(s)
- Jung Yeon Lee
- Department of Biochemistry and Molecular Biology, School of Medicine, Medical Research Center for Bioreaction to Reactive Oxygen Species, Biomedical Science Institute, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
15
|
Wong VKW, Zhou H, Cheung SSF, Li T, Liu L. Mechanistic study of saikosaponin-d (Ssd) on suppression of murine T lymphocyte activation. J Cell Biochem 2009; 107:303-15. [PMID: 19301261 DOI: 10.1002/jcb.22126] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Saikosaponin-d (Ssd) is a triterpene saponin derived from the medicinal plant, Bupleurum falcatum L. (Umbelliferae). Previous findings showed that Ssd exhibits a variety of pharmacological and immunomodulatory activities including anti-inflammatory, anti-bacterial, anti-viral and anti-cancer effects. In the current study we have investigated the effects of Ssd on activated mouse T lymphocytes through the NF-kappaB, NF-AT and AP-1 signaling pathways, cytokine secretion, and IL-2 receptor expression. The results demonstrated that Ssd not only suppressed OKT3/CD28-costimulated human T cell proliferation, it also inhibited PMA, PMA/Ionomycin and Con A-induced mouse T cell activation in vitro. The inhibitory effect of Ssd on PMA-induced T cell activation was associated with down-regulation of NF-kappaB signaling through suppression of IKK and Akt activities. In addition, Ssd suppressed both DNA binding activity and the nuclear translocation of NF-AT and activator protein 1 (AP-1) of the PMA/Ionomycin-stimulated T cells. The cell surface markers like IL-2 receptor (CD25) were also down-regulated together with decreased production of pro-inflammatory cytokines of IL-6, TNF-alpha and IFN-gamma. These results indicate that the NF-kappaB, NF-AT and AP-1 (c-Fos) signaling pathways are involved in the T cell inhibition evoked by Ssd, so it can be a potential candidate for further study in treating T cell-mediated autoimmune conditions.
Collapse
|
16
|
Higai K, Tsukada M, Moriya Y, Azuma Y, Matsumoto K. Prolonged high glucose suppresses phorbol 12-myristate 13-acetate and ionomycin-induced interleukin-2 mRNA expression in Jurkat cells. Biochim Biophys Acta Gen Subj 2008; 1790:8-15. [PMID: 18992303 DOI: 10.1016/j.bbagen.2008.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 09/29/2008] [Accepted: 10/03/2008] [Indexed: 01/13/2023]
Abstract
BACKGROUND The disturbance of immunological responses is a complication of diabetes mellitus. METHODS AND RESULTS We cultured Jurkat cells in 11.1 (normal) and 22.2 mmol/l (high) glucose for 12 weeks and stimulated them with 10 nmol/l phorbol 12-myristate 13-acetate (PMA) and 500 nmol/l ionomycin. RT-PCR revealed that induced interleukin (IL)-2 mRNA expression levels were suppressed in high glucose cultures compared to those in normal glucose. Promoter activities of IL-2, nuclear factor of activated T cells (NFAT), and activator protein-1 (AP-1), after 6 h stimulation with PMA and ionomycin, gradually decreased in high glucose cultures to approximately 20% of those in normal glucose at 12 weeks. The prolonged culture in high glucose increased inducible cAMP early repressor (ICER) II mRNA and protein levels, and overexpression of ICER II dose-dependently suppressed promoter activities of IL-2, NFAT, and AP-1. Moreover, ICER II mRNA expression was transiently induced by stimulation with PMA and ionomycin in normal glucose cultures; however, with high glucose, the induction disappeared. CONCLUSION These results indicate that ICER II protein accumulates during prolonged culture in high glucose and suppresses IL-2 mRNA expression in Jurkat cells.
Collapse
Affiliation(s)
- Koji Higai
- Department of Clinical Chemistry, School of Pharmaceutical Sciences, Toho University Miyama 2-2-1, Funabashi, Chiba 247-8510, Japan.
| | | | | | | | | |
Collapse
|
17
|
Yoon H, Oh YT, Lee JY, Choi JH, Lee JH, Baik HH, Kim SS, Choe W, Yoon KS, Ha J, Kang I. Activation of AMP-activated protein kinase by kainic acid mediates brain-derived neurotrophic factor expression through a NF-kappaB dependent mechanism in C6 glioma cells. Biochem Biophys Res Commun 2008; 371:495-500. [PMID: 18445478 DOI: 10.1016/j.bbrc.2008.04.102] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Accepted: 04/19/2008] [Indexed: 12/12/2022]
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of energy homeostasis. Kainic acid (KA), a prototype excitotoxin is known to induce brain-derived neurotrophic factor (BDNF) in brain. In this study, we examined the role of AMPK in KA-induced BDNF expression in C6 glioma cells. We showed that KA and KA receptor agonist induced activation of AMPK and KA-induced AMPK activation was blocked by inhibition of Ca(2+)/calmodulin-dependent protein kinase kinase (CaMKK) beta. We then showed that inhibition of AMPK by compound C, a selective inhibitor of AMPK, or small interfering RNA of AMPKalpha1 blocked KA-induced BDNF mRNA and protein expression. Inhibition of AMPK blocked KA-induced phosphorylation of CaMKII and I kappaB kinase (IKK) in C6 cells. Finally, we showed that inhibition of AMPK reduced DNA binding and transcriptional activation of nuclear factor-kappaB (NF-kappaB) in KA-treated cells. These results suggest that AMPK mediates KA-induced BDNF expression by regulating NF-kappaB activation.
Collapse
Affiliation(s)
- Hana Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Oh YT, Lee JY, Yoon H, Lee EH, Baik HH, Kim SS, Ha J, Yoon KS, Choe W, Kang I. Lipopolysaccharide induces hypoxia-inducible factor-1 alpha mRNA expression and activation via NADPH oxidase and Sp1-dependent pathway in BV2 murine microglial cells. Neurosci Lett 2007; 431:155-60. [PMID: 18164813 DOI: 10.1016/j.neulet.2007.11.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 11/06/2007] [Accepted: 11/16/2007] [Indexed: 02/05/2023]
Abstract
Hypoxia-inducible factor-1 (HIF-1), the key transcription factor of hypoxia-inducible genes, is known to be involved in inflammation and immune response, but little is known about the regulation of HIF-1 during microglial activation. Thus, we examined effect of lipopolysaccharide (LPS) on HIF-1 activation and its signaling mechanism in BV2 microglial cells. LPS induced HIF-1alpha mRNA and protein expression as well as HIF-1 transcriptional activation. Moreover, HIF-1alpha knockdown by small interfering RNA (siRNA) decreased LPS-induced expression of hypoxia responsive genes, VEGF, iNOS, and COX-2. We then showed that LPS-induced HIF-1alpha mRNA expression was blocked by an antioxidant, NADPH oxidase inhibitors, and siRNA of gp91phox, a subunit of NADPH oxidase. In addition, we showed that specific pharmacological inhibitors of PI 3-kinase and protein kinase C decreased LPS-induced HIF-1alpha mRNA expression. Finally, we showed that inhibition of transcription factor Sp1 by mithramycin A or Sp1 siRNA decreased LPS-induced HIF-1alpha mRNA and protein expression. Consistently, LPS increased Sp1 DNA binding and its transcriptional activity. Taken together, these results suggest that LPS induces HIF-1alpha mRNA expression and activation via NADPH oxidase and Sp1 in BV2 microglia.
Collapse
Affiliation(s)
- Young Taek Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The role of the paxillin superfamily of adaptor proteins in B cell antigen receptor (BCR) signaling has not been studied previously. We show here that leupaxin (LPXN), a member of this family, was tyrosine-phosphorylated and recruited to the plasma membrane of human BJAB lymphoma cells upon BCR stimulation and that it interacted with Lyn (a critical Src family tyrosine kinase in BCR signaling) in a BCR-induced manner. LPXN contains four leucine-rich sequences termed LD motifs, and serial truncation and specific domain deletion of LPXN indicated that its LD3 domain is involved in the binding of Lyn. Of a total of 11 tyrosine sites in LPXN, we mutated Tyr(22), Tyr(72), Tyr(198), and Tyr(257) to phenylalanine and demonstrated that LPXN was phosphorylated by Lyn only at Tyr(72) and that this tyrosine site is proximal to the LD3 domain. The overexpression of LPXN in mouse A20 B lymphoma cells led to the suppression of BCR-induced activation of JNK, p38 MAPK, and, to a lesser extent, Akt, but not ERK and NFkappaB, suggesting that LPXN can selectively repress BCR signaling. We further show that LPXN suppressed the secretion of interleukin-2 by BCR-activated A20 B cells and that this inhibition was abrogated in the Y72F LPXN mutant, indicating that the phosphorylation of Tyr(72) is critical for the biological function of LPXN. Thus, LPXN plays an inhibitory role in BCR signaling and B cell function.
Collapse
Affiliation(s)
- Valerie Chew
- Laboratory of Immune Regulation, Biomedical Sciences Institutes, Agency for Science, Technology and Research and Singapore Immunology Network, Singapore 138673, Singapore
| | - Kong-Peng Lam
- Laboratory of Immune Regulation, Biomedical Sciences Institutes, Agency for Science, Technology and Research and Singapore Immunology Network, Singapore 138673, Singapore.
| |
Collapse
|