1
|
Huang X, Yuan J, Gu J, Abbas Y, Yuan Y, Liu Z, Zou H, Bian J. Protective effect of honokiol on cadmium-induced liver injury in chickens. Poult Sci 2024; 103:104066. [PMID: 39067123 PMCID: PMC11338095 DOI: 10.1016/j.psj.2024.104066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Cadmium (Cd), a highly toxic heavy metal in the environment, poses a significant threat to livestock and poultry farming. Honokiol (HNK), a Chinese herbal extract with potent antioxidant activity, acts through oxidative damage and inflammation. Cd induces oxidative stress and causes liver damage in animals. However, whether HNK can alleviate Cd-induced liver injury in chickens and its mechanism remains unclear. In this study, the 48 chickens were randomly allocated into 4 groups, control group, Cd group (70 mg/kg Cd), HNK group (200 mg/kg HNK) and Cd + HNK group (70 mg/kg Cd+200 mg/kg HNK). Results showed that HNK improved the Cd induced reduction in chicken body weight, liver weight, and liver coefficient. HNK recovered the Cd induced liver damaged through increased serum liver biochemical indexes, impaired liver oxidase activity and the disordered the expression level of antioxidant genes. HNK alleviated Cd induced pathological and ultrastructure damage of liver tissue and liver cell that leads apoptosis. HNK decreased Cd contents in the liver, Cd induced disturbances in the levels of trace elements such as iron, copper, zinc, manganese, and selenium. HNK attenuated the damage to the gap junction structure of chicken liver cells caused by Cd and reduced the impairment of oxidase activity and the expression level of antioxidant genes induced by Cd. In conclusion, HNK presents essential preventive measures and a novel pharmacological potential therapy against Cd induced liver injury. Our experiments show that HNK can be used as a new green feed additive in the poultry industry, which provides a theoretical basis for HNK to deal with the pollution caused by Cd in the poultry industry.
Collapse
Affiliation(s)
- Xiaoqian Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Junzhao Yuan
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450000, Henan, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yassar Abbas
- Department of Animal Sciences, Jhang Campus, University of Veterinary and Animal Sciences, Jhang 54590, Pakistan
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
2
|
Wang M, Zhou Y, Jian Q, Ai Z, Zhou S. Mechanisms of Rostellularia procumbens (L.) Nees on treating chronic glomerulonephritis explored by network pharmacology, RNA-seq, and in vitro experiments. BMC Complement Med Ther 2023; 23:263. [PMID: 37488573 PMCID: PMC10367255 DOI: 10.1186/s12906-023-04079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND The purpose of this study was to demonstrate the in vitro anti-nephritis activity of Rostellularia procumbens (L.) Nees (R. procumbens) extract and to make a preliminary investigation of its anti-nephritis mechanism. METHODS A prediction network was built that describes the relationship between R. procumbens and CGN. Then, the potential targets for R. procumbens against CGN were imported into the DAVID database for Gene Ontology (GO) biological annotation analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. A lipopolysaccharide (LPS)-stimulated rat mesangial cell HBZY-1 model in vitro was used to examine the anti-inflammatory activity of R. procumbens extract. RNA-seq was utilized to investigate differentially expressed genes (DEGs) and enriched signaling pathways between groups. Finally, qPCR was used for the validation analysis of the experimental results. RESULTS The results of network pharmacology showed that R. procumbens exerts its therapeutic effect on CGN through the AGE-RAGE signaling pathway in diabetic complications, PI3K-Akt, IL-17 signaling pathway, and so on. R. procumbens n-butanol extract (J-NE) can effectively relieve inflammation in HBZY-1. The results of KEGG pathway enrichment suggest that J-NE attenuated CGN was associated with the IL-17 signaling pathway, and the results of RNA-seq were consistent with network pharmacology. Targets enriched in the IL-17 signaling pathway, including Chemokine (C-C motif) ligand 7 (CCL7), Lipocalin 2 (LCN2), Chemokine (C-C motif) ligand 2 (CCL2), and Chemokine (C-X-C motif) ligand 1 (CXCL1), have been identified as crucial targets attenuating CGN by J-NE. CONCLUSION R. procumbens is a promising pharmacological candidate for the treatment of CGN in the present era.
Collapse
Affiliation(s)
- Mengfan Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Huangjiahu Road (West), Hongshan District, Wuhan, Hubei Province, 430065, China
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Huangjiahu Road (West), Hongshan District, Wuhan, Hubei Province, 430065, China
| | - Qiuyuan Jian
- School of Pharmacy, Hubei University of Chinese Medicine, Huangjiahu Road (West), Hongshan District, Wuhan, Hubei Province, 430065, China
| | - Zhongzhu Ai
- School of Pharmacy, Hubei University of Chinese Medicine, Huangjiahu Road (West), Hongshan District, Wuhan, Hubei Province, 430065, China.
- Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Hubei University of Chinese Medicine, Wuhan, 430065, China.
- Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Shanshan Zhou
- The First Clinical Medical School, Hubei University of Chinese Medicine, Huangjiahu Road (West), Hongshan District, Wuhan, Hubei Province, 430065, China.
| |
Collapse
|
3
|
Gilham D, Wasiak S, Rakai BD, Fu L, Tsujikawa LM, Sarsons CD, Carestia A, Lebioda K, Johansson JO, Sweeney M, Kalantar-Zadeh K, Kulikowski E. Apabetalone Downregulates Fibrotic, Inflammatory and Calcific Processes in Renal Mesangial Cells and Patients with Renal Impairment. Biomedicines 2023; 11:1663. [PMID: 37371758 DOI: 10.3390/biomedicines11061663] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Epigenetic mechanisms are implicated in transcriptional programs driving chronic kidney disease (CKD). Apabetalone is an orally available inhibitor of bromodomain and extraterminal (BET) proteins, which are epigenetic readers that modulate gene expression. In the phase 3 BETonMACE trial, apabetalone reduced risk of major adverse cardiac events (MACE) by 50% in the CKD subpopulation, indicating favorable effects along the kidney-heart axis. Activation of human renal mesangial cells (HRMCs) to a contractile phenotype that overproduces extracellular matrix (ECM) and inflammatory cytokines, and promotes calcification, frequently accompanies CKD to drive pathology. Here, we show apabetalone downregulated HRMC activation with TGF-β1 stimulation by suppressing TGF-β1-induced α-smooth muscle actin (α-SMA) expression, α-SMA assembly into stress fibers, enhanced contraction, collagen overproduction, and expression of key drivers of fibrosis, inflammation, or calcification including thrombospondin, fibronectin, periostin, SPARC, interleukin 6, and alkaline phosphatase. Lipopolysaccharide-stimulated expression of inflammatory genes IL6, IL1B, and PTGS2 was also suppressed. Transcriptomics confirmed apabetalone affected gene sets of ECM remodeling and integrins. Clinical translation of in vitro results was indicated in CKD patients where a single dose of apabetalone reduced plasma levels of key pro-fibrotic and inflammatory markers, and indicated inhibition of TGF-β1 signaling. While plasma proteins cannot be traced to the kidney alone, anti-fibrotic and anti-inflammatory effects of apabetalone identified in this study are consistent with the observed decrease in cardiovascular risk in CKD patients.
Collapse
Affiliation(s)
- Dean Gilham
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Sylwia Wasiak
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Brooke D Rakai
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Li Fu
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Laura M Tsujikawa
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | | | - Agostina Carestia
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Kenneth Lebioda
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| | - Jan O Johansson
- Resverlogix Inc., 535 Mission St, 14th Floor, San Francisco, CA 94105, USA
| | - Michael Sweeney
- Resverlogix Inc., 535 Mission St, 14th Floor, San Francisco, CA 94105, USA
| | - Kamyar Kalantar-Zadeh
- Harbor-UCLA Medical Center, University of California Los Angeles, 1000 W Carson St, Torrance, CA 90502, USA
| | - Ewelina Kulikowski
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB T3E 6L1, Canada
| |
Collapse
|
4
|
Cytoprotective remedies for ameliorating nephrotoxicity induced by renal oxidative stress. Life Sci 2023; 318:121466. [PMID: 36773693 DOI: 10.1016/j.lfs.2023.121466] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
AIMS Nephrotoxicity is the hallmark of anti-neoplastic drug metabolism that causes oxidative stress. External chemical agents and prescription drugs release copious amounts of free radicals originating from molecular oxidation and unless sustainably scavenged, they stimulate membrane lipid peroxidation and disruption of the host antioxidant mechanisms. This review aims to provide a comprehensive collection of potential cytoprotective remedies in surmounting the most difficult aspect of cancer therapy as well as preventing renal oxidative stress by other means. MATERIALS AND METHODS Over 400 published research and review articles spanning several decades were scrutinised to obtain the relevant data which is presented in 3 categories; sources, mechanisms, and mitigation of renal oxidative stress. KEY-FINDINGS Drug and chemical-induced nephrotoxicity commonly manifests as chronic or acute kidney disease, nephritis, nephrotic syndrome, and nephrosis. Renal replacement therapy requirements and mortalities from end-stage renal disease are set to rapidly increase in the next decade for which 43 different cytoprotective compounds which have the capability to suppress experimental nephrotoxicity are described. SIGNIFICANCE The renal system performs essential homeostatic functions that play a significant role in eliminating toxicants, and its accumulation and recurrence in nephric tissues results in tubular degeneration and subsequent renal impairment. Global statistics of the latest chronic kidney disease prevalence is 13.4 % while the end-stage kidney disease requiring renal replacement therapy is 4-7 million per annum. The remedial compounds discussed herein had proven efficacy against nephrotoxicity manifested consequent to impaired antioxidant mechanisms in preclinical models produced by renal oxidative stress activators.
Collapse
|
5
|
Ding Q, Wang Y, Yang C, Li X, Yu X. Clinical Utility of the Sivelestat for the Treatment of ALI/ARDS: Moving on in the Controversy? INTENSIVE CARE RESEARCH 2023; 3:12-17. [PMID: 36911225 PMCID: PMC9412801 DOI: 10.1007/s44231-022-00012-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 01/08/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a serious condition that can arise following direct or indirect acute lung injury (ALI). It is heterogeneous and has a high mortality rate. Supportive care is the mainstay of treatment and there is no definitive pharmacological treatment as yet. In nonclinical studies, neutrophil elastase inhibitor sivelestat appears to show benefit in ARDS without inhibiting the host immune defense in cases of infection. In clinical studies, the efficacy of sivelestat in the treatment of ARDS remains controversial. The currently available evidence suggests that sivelestat may show some benefit in the treatment of ARDS, although large, randomized controlled trials are needed in specific pathophysiological conditions to explore these potential benefits.
Collapse
Affiliation(s)
- Qiongli Ding
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Yi Wang
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Chunbo Yang
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Xiang Li
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| | - Xiangyou Yu
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Medical University, Urumqi, 830054 Xinjiang Uygur Autonomous Region China.,Xinjiang Uygur Autonomous Region Institute of Critical Medicine, Urumqi, 830054 Xinjiang Uygur Autonomous Region China
| |
Collapse
|
6
|
Zhong Z, Guo X, Zheng Y. Network Pharmacology-Based and Molecular Docking Analysis of Resveratrol's Pharmacological Effects on Type I Endometrial Cancer. Anticancer Agents Med Chem 2022; 22:1933-1944. [PMID: 34773964 PMCID: PMC9241081 DOI: 10.2174/1871520621666211015140455] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/19/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Resveratrol is a natural polyphenol commonly seen in foods. It has demonstrated an inhibitive effect on endometrial cancer, but the molecular action is still not known. OBJECTIVE We aimed to use network pharmacology to systematically study the possible mechanisms of resveratrol's pharmacological effects on type I endometrial cancer. METHODS Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) were used to predict resveratrol's possible target genes. They were then converted to UniProt gene symbols. Simultaneously, type I endometrial cancer-related target genes were collected from GeneCards. All data were pooled to identify common target genes. The protein-protein interaction (PPI) network was constructed and further analyzed via STRING Online Database. Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were also performed afterward. To visualise resveratrol's overall pharmacological effects on type I endometrial cancer, a network of drug components-target gene-disease (CTD) was constructed. Then, we performed in silico molecular docking study to validate the possible binding conformation between resveratrol and candidate targets. RESULTS There are 150 target genes of resveratrol retrieved after UniProt conversion; 122 of them shared interaction with type I endometrial cancer. Some important oncogenes and signaling pathways are involved in the process of resveratrol's pharmacological effects on endometrioid cancer. Molecular docking analysis confirmed that hydrogen bonding and hydrophobic interaction are the main interaction between resveratrol and its targets. CONCLUSION We have explored the possible underlying mechanism of resveratrol in antagonising type I endometrial cancer through a network pharmacology-based approach and in-silico verification. However, further experiments are necessary to add to the evidence identifying resveratrol as a promising anti-type I endometrial cancer agent.
Collapse
Affiliation(s)
- Zixing Zhong
- Department of Obstetrics, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou310014, China
| | - Xin Guo
- Department of Obstetrics, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou310014, China
| | - Yanmei Zheng
- Department of Obstetrics, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou310014, China
| |
Collapse
|
7
|
Howell S, Song W, Pastuszak A, Khera M. Differential Gene Expression in Post-Finasteride Syndrome Patients. J Sex Med 2021; 18:1479-1490. [PMID: 37057444 DOI: 10.1016/j.jsxm.2021.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/28/2021] [Accepted: 05/14/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND An organic etiology underpinning post-finasteride syndrome, a constellation of persistent sexual, neuropsychiatric, and somatic symptoms reported by men exposed to 5-alpha-reductase inhibitors (5ARIs), is debated. Persistent changes in neurosteroid levels or androgen receptor expression have been implicated. AIM To determine whether differences in gene expression, especially in relevant biologic pathways, exist between patients reporting post-finasteride syndrome symptoms and healthy controls. METHODS This was a single center, prospective case-control study taking place between March 2013 and September 2018. Men 18 years and older being evaluated for sexual dysfunction (study) or circumcision (control) were eligible for inclusion. Twenty-six men with a history of 5ARI use reporting symptoms consistent with post-finasteride syndrome were included in the patient group. Twenty-six men consented to inclusion in the control group. OUTCOMES The primary outcome measure is gene expression data for genes affecting neurosteroid levels and androgen receptor activity from penile skin cells. RESULTS Gene expression of cells from penile skin samples from twenty-six men of median age 38 years (IQR, 33-42) in the study group was compared with that from twenty-six men of median age 41 years (IQR, 35-62) in the control group (P = .13), with 1,446 genes significantly over-expressed and 2,318 genes significantly under-expressed in study patients. Androgen receptor expression was significantly higher in study patients compared to controls (9.961 vs 9.494, adjusted P value = .01). Serum levels of androgen receptor activity markers 5α-androstanediol (0.950 ng/mL [0.749-1.587] vs 0.949 [0.817-1.337], P = .34) or 3α-androstanedione (3.1 ng/mL [1.925-5.475] vs 6.7 [3.375-11.4], P = .31) revealed no significant differences. No significant differences were found between the number of trinucleotide repeats (21.5 [20-23.75], 22 [19-25], P = .94). CLINICAL IMPLICATIONS In this study we present evidence of gene expression correlating with observed biologic differences in patients with post-finasteride syndrome; providers who prescribe 5ARIs should be aware and advise their patients accordingly. STRENGTHS & LIMITATIONS Strengths of this study include the evaluation of multiple proposed etiologies for post-finasteride syndrome. The study is also strengthened by the fact that not all data matched the initial hypotheses, qualifying the argument for the existence of PFS. Limitations include potential selection bias arising from more severe phenotypes seeking care; lack of gene expression data prior to 5ARI exposure; lack of non-penile tissue samples supposedly involved; and a lack of mechanistic data to imply causality. CONCLUSION This study is the first to consider and demonstrate gene expression differences in patients with PFS as a potential etiology of sexual dysfunction. Howell S, Song W, Pastuszak A, et al. Differential Gene Expression in Post-Finasteride Syndrome Patients. J Sex Med 2021;18:1479-1490.
Collapse
Affiliation(s)
- Skyler Howell
- Division of Urology, Department of Surgery, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Weitao Song
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Alexander Pastuszak
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mohit Khera
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Chen C, Zhang QW, Ye Y, Lin LG. Honokiol: A naturally occurring lignan with pleiotropic bioactivities. Chin J Nat Med 2021; 19:481-490. [PMID: 34247771 DOI: 10.1016/s1875-5364(21)60047-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Indexed: 12/16/2022]
Abstract
Honokiol is the dominant biphenolic compound isolated from the Magnolia tree, and has long been considered as the active constituent of the traditional Chinese herb, 'Houpo', which is widely used to treat symptoms due to 'stagnation of qi'. Pharmacological studies have shown that honokiol possesses a wide range of bioactivities without obvious toxicity. Honokiol protects the liver, kidneys, nervous system, and cardiovascular system through reducing oxidative stress and relieving inflammation. Moreover, honokiol shows anti-diabetic property through enhancing insulin sensitivity, and anti-obese property through promoting browning of adipocytes. In vivo and in vitro studies indicated that honokiol functions as an anti-cancer agent through multiple mechanisms: inhibiting angiogenesis, promoting cell apoptosis, and regulating cell cycle. A variety of therapeutic effects of honokiol may be associated with its physiochemical properties, which make honokiol readily cross the blood brain barrier and the blood-cerebrospinal fluid barrier, with high bioavailability. In the future, more clinical researches on honokiol are needed to fully authenticate its therapeutic values.
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yang Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
9
|
Li Z, Guo H, Li J, Ma T, Zhou S, Zhang Z, Miao L, Cai L. Sulforaphane prevents type 2 diabetes-induced nephropathy via AMPK-mediated activation of lipid metabolic pathways and Nrf2 antioxidative function. Clin Sci (Lond) 2020; 134:2469-2487. [PMID: 32940670 DOI: 10.1042/cs20191088] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
Sulforaphane (SFN) prevents diabetic nephropathy (DN) in type 2 diabetes (T2D) by up-regulating nuclear factor (erythroid-derived 2)-like 2 (Nrf2). AMP-activated protein kinase (AMPK) can attenuate the pathogenesis of DN by improving renal lipotoxicity along with the activation of Nrf2-mediated antioxidative signaling. Therefore, we investigated whether AMPKα2, the central subunit of AMPK in energy metabolism, is required for SFN protection against DN in T2D, and whether potential cross-talk occurs between AMPKα2 and Nrf2. AMPKα2 knockout (Ampkα2-/-) mice and wildtype (WT) mice were fed a high-fat diet (HFD) or a normal diet (ND) to induce insulin resistance, followed by streptozotocin (STZ) injection to induce hyperglycemia, as a T2D model. Both T2D and control mice were treated with SFN or vehicle for 3 months. At the end of the 3-month treatment, all mice were maintained only on HFD or ND for an additional 3 months without SFN treatment. Mice were killed at sixth month after T2D onset. Twenty-four-hour urine albumin at third and sixth months was significantly increased as renal dysfunction, along with significant renal pathological changes and biochemical changes including renal hypertrophy, oxidative damage, inflammation, and fibrosis in WT T2D mice, which were prevented by SFN in certain contexts, but not in Ampkα2-/- T2D mice. SFN prevention of T2D-induced renal lipotoxicity was associated with AMPK-mediated activation of lipid metabolism and Nrf2-dependent antioxidative function in WT mice, but not in SFN-treated Ampkα2-/- mice. Therefore, SFN prevention of DN is AMPKα2-mediated activation of probably both lipid metabolism and Nrf2 via AMPK/AKT/glycogen synthase kinase (GSK)-3β/Src family tyrosine kinase (Fyn) pathways.
Collapse
Affiliation(s)
- Zhuo Li
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Hua Guo
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Department of Immunology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jia Li
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Departments of Cardiology and Nephrology at The First Hospital of Jilin University, Changchun 130021, China
| | - Tianjiao Ma
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Shanshan Zhou
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Departments of Cardiology and Nephrology at The First Hospital of Jilin University, Changchun 130021, China
| | - Zhiguo Zhang
- Departments of Cardiology and Nephrology at The First Hospital of Jilin University, Changchun 130021, China
| | - Lining Miao
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, Norton Children and University of Louisville School of Medicine, Louisville 40202, KY, U.S.A
- Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville 40202, KY, U.S.A
| |
Collapse
|
10
|
Zhang LH, Xiao B, Zhong M, Li Q, Chen JY, Huang JR, Rao H. LncRNA NEAT1 accelerates renal mesangial cell injury via modulating the miR-146b/TRAF6/NF-κB axis in lupus nephritis. Cell Tissue Res 2020; 382:627-638. [PMID: 32710276 DOI: 10.1007/s00441-020-03248-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/22/2020] [Indexed: 12/31/2022]
Abstract
Although growing advances have been made in the regulation of lupus nephritis recently, lupus nephritis is still one of the major causes of death in SLE patients and the pathogenesis remains largely unknown. Therefore, exploring the pathological mechanisms is urgently needed for designing and developing novel therapeutic strategies for lupus nephritis. Human renal mesangial cells (HRMCs) were transfected with sh-NEAT1, miR-146b mimic, pcDNA-NEAT1, miR-146b inhibitor, or sh-TRAF6 to modify their expression. Lipopolysaccharide (LPS) was used to induce inflammatory injury. Cell viability was examined with CCK8. Apoptosis was determined by flow cytometry and Hoechst staining. qRT-PCR and western blot were used to analyze gene expression. The secretion of inflammatory cytokines was examined with ELISA. The bindings of NEAT1 with miR-146b and miR-146b with TRAF6 were tested by dual-luciferase reporter assay. NEAT1 was upregulated in LPS-treated HRMCs. Both the knockdown of NEAT1 and TRAF6 suppressed the LPS-induced inflammatory injury in HRMCs. NEAT1 directly targeted miR-146b to control miR-146b-mediated regulation of TRAF6 expression in HRMCs. NEAT1 promoted the expression of TRAF6 via targeting miR-146b to accelerate the LPS-mediated renal mesangial cell injury in HRMCs. Moreover, TRAF6 activated the NF-κB signaling in HRMCs. NEAT1 accelerated renal mesangial cell injury via directly targeting miR-146b, promoting the expression of TRAF6, and activating the NF-κB signaling in lupus nephritis. Our investigation elucidated novel pathological mechanisms and provided potential therapeutic targets for lupus nephritis.
Collapse
Affiliation(s)
- Li-Hua Zhang
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Bin Xiao
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Miao Zhong
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Qiao Li
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Jian-Ying Chen
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Jie-Rou Huang
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China
| | - Hui Rao
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No.89, Guhan Road, Furong District, Changsha, 410016, Hunan Province, People's Republic of China.
| |
Collapse
|
11
|
Gold Clusters Attenuate Inflammation in Rat Mesangial Cells via Inhibiting the Activation of NF-κB Pathway. NANOMATERIALS 2020; 10:nano10040712. [PMID: 32290032 PMCID: PMC7221671 DOI: 10.3390/nano10040712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
Sepsis-induced acute kidney injury (AKI) with high incidence and mortality rates remains a great challenge in the clinic; thus, novel therapies need to be developed urgently. This complication is associated with an overwhelming systemic inflammatory response. The aim of this study was to evaluate the potential effects and possible mechanisms of gold clusters on septic AKI in vitro. Rat mesangial HBZY-1 cells were treated with peptide-templated gold clusters under lipopolysaccharide (LPS) stimulation. The LPS-induced expression of pro-inflammatory cytokines was measured, including tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β) and interleukin-6 (IL-6). Our data showed that the LPS-induced transcription and secretion of these cytokines were suppressed by pretreatment of gold clusters in a dose-dependent manner. Cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) also play key roles in septic AKI and both of them are induced upon LPS-stimulation in mesangial cells. Our results further showed that pretreatment with gold clusters dramatically inhibited the LPS-stimulated transcription and expression of COX2 and iNOS, and the subsequent prostaglandin E2 (PGE2) and nitric oxide (NO) production in HBZY-1 cells. Since these factors are involved in the NF-κB pathway upon LPS stimulation, the potential roles of gold clusters on the NF-κB pathway were further determined. We found that LPS-induced NF-κB activation was suppressed in gold clusters-pretreated HBZY-1 cells. These results demonstrated that gold clusters can attenuate LPS-induced inflammation in mesangial cells, probably via inhibiting the activation of the NF-κB pathway, suggesting a potential therapeutic approach for septic AKI.
Collapse
|
12
|
Elfeky MG, Mantawy EM, Gad AM, Fawzy HM, El-Demerdash E. Mechanistic aspects of antifibrotic effects of honokiol in Con A-induced liver fibrosis in rats: Emphasis on TGF-β/SMAD/MAPK signaling pathways. Life Sci 2020; 240:117096. [PMID: 31760097 DOI: 10.1016/j.lfs.2019.117096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023]
Abstract
UNLABELLED Aim Liver fibrosis represents a massive global health burden with limited therapeutic options. Thus, the need for curative options is evident. Thus, this study aimed to assess the potential antifibrotic effect of honokiol in Concanavalin A (Con A) induced immunological model of liver fibrosis as well the possible underlying molecular mechanisms. METHODS Male Sprague-Dawley rats were treated with either Con A (20 mg/kg, IV) and/or honokiol (10 mg/kg, orally) for 4 weeks. Hepatotoxicity indices were as well as histopathological evaluation was done. Hepatic fibrosis was assessed by measuring alpha smooth muscle actin (α-SMA) expression and collagen fibers deposition by Masson's trichrome stain and hydroxyproline content. To elucidate the underlying molecular mechanisms, the effect of honokiol on oxidative stress, inflammatory markers as well as transforming growth factor beta (TGF-β)/SMAD and mitogen-activated protein kinase (MAPK) pathways was assessed. KEY FINDINGS Honokiol effectively reversed the hepatotoxicity indices elevations and abnormal histopathological changes induced by Con A. Besides, honokiol attenuated Con A-induced liver fibrosis by down-regulation of hydroxyproline levels, α-SMA expression together with a marked decrease in collagen fibers deposition. Mechanistically Con A induced oxidative stress, provocation of inflammatory responses and activation of TGF-β/SMAD/MAPK pathways. Contrariwise, honokiol co-treatment significantly restored antioxidant defence mechanisms, down-regulated inflammatory cascades and inhibited TGF-β/SMAD/MAPK signaling pathways. CONCLUSION The results provide an evidence for the promising antifibrotic effect of honokiol that could be partially due to suppressing oxidative stress and inflammatory processes as well as inhibition of TGF-β/SMAD/MAPK signaling pathways.
Collapse
Affiliation(s)
- Maha G Elfeky
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Hala M Fawzy
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
13
|
Zhang T, Xiang L. Honokiol alleviates sepsis-induced acute kidney injury in mice by targeting the miR-218-5p/heme oxygenase-1 signaling pathway. Cell Mol Biol Lett 2019; 24:15. [PMID: 30833971 PMCID: PMC6387556 DOI: 10.1186/s11658-019-0142-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/13/2019] [Indexed: 12/30/2022] Open
Abstract
Background Honokiol is a low-molecular-weight natural product and has been reported to exhibit anti-inflammatory activity. Objectives Our study aimed to investigate the influence of honokiol on sepsis-induced acute kidney injury (AKI) in a mouse model. Material and methods A cecal ligation and puncture (CLP) surgical operation was performed to establish a sepsis-induced acute kidney injury model in mice. Renal histomorphological analysis was performed with periodic acid-Schiff (PAS) staining. The levels of inflammatory markers in serum were measured by ELISA assay. The mRNA and protein levels were assayed by RT-qPCR and western blotting, respectively. Annexin V-FITC/PI staining was used to evaluate glomerular mesangial cell (GMC) apoptosis. Results The results revealed that honokiol significantly increased the survival rate in mice undergoing a CLP operation. Inflammatory cytokines, such as TNF-α, IL-6 and IL-1β, were significantly inhibited in honokiol-treated septic mice compared with the CLP group. In addition, honokiol showed the ability to reverse CLP-induced AKI in septic mice. Furthermore, heme oxygenase-1 (HO-1) expression levels were significantly up-regulated and miR-218-5p was markedly down-regulated in honokiol-treated septic mice as compared to CLP-operated mice. Bioinformatics and experimental measurements showed that HO-1 was a direct target of miR-218-5p. In vitro experiments showed that both honokiol and miR-218-5p inhibitors blocked lipopolysaccharide (LPS)-induced cell growth inhibition and GMC apoptosis by increasing the expression of HO-1. Conclusions Honokiol ameliorated AKI in septic mice and LPS-induced GMC dysfunction, and the underlying mechanism was mediated, at least partially, through the regulation of miR-218-5p/HO-1 signaling. Electronic supplementary material The online version of this article (10.1186/s11658-019-0142-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tao Zhang
- 1Department of of Intensive Care Unit, Tianjin Huanhu Hospital, No. 6 Jizhao Road, Tianjin, 300060 People's Republic of China
| | - Lei Xiang
- 2Department of Neurology, Tianjin Huanhu Hospital, Tianjin, 300060 People's Republic of China
| |
Collapse
|
14
|
Wu F, Yao H, Zheng F, Tang S, Lin X, Li L, Zhou J, Li H. Protective effects of honokiol against oxidative stress-induced apoptotic signaling in mouse podocytes treated with H2O2. Exp Ther Med 2018; 16:1278-1284. [PMID: 30116378 PMCID: PMC6090302 DOI: 10.3892/etm.2018.6313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/13/2018] [Indexed: 01/11/2023] Open
Abstract
Honokiol (HNK), an important bioactive compound purified from Magnolia officinalis Cortex, has been demonstrated to have manifold beneficial anti-oxidative, anti-inflammatory, anti-bacterial and antitumor pharmacological effects. In the present study, the association of HNK in the signaling mechanism associated with hydrogen peroxide (H2O2)-induced apoptosis of cultured mouse podocytes was investigated. HNK did not cause significant changes in podocyte viability when its concentration remained below 20 µM. MTS assay and flow cytometry confirmed that H2O2 significantly enhanced the rates of apoptosis while produce significant reduction in viability of podocytes. Following 24 h of pre-treatment with different concentrations of HNK, the viability of adherent podocytes increased and apoptosis significantly decreased in a dose-dependent manner below 20 µM. Reverse transcription-polymerase chain reaction and western blot results indicated that HNK significantly decreased the expression of mRNA and cleaved protein of caspase-3 and caspase-9 in podocytes pre-treated with H2O2. Furthermore, phosphorylation of the signaling molecules protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) 1/2 appeared to increase following HNK treatment. In conclusion, HNK largely eliminated the role of promoting podocyte apoptosis in an oxidative stress environment, which was a protective factor on podocytes cultured with H2O2. The anti-oxidative stress mechanisms of HNK are partly due to suppressing the expression of caspase-3 and caspase-9 and upregulating phosphorylated-Akt and -Erk 1/2.
Collapse
Affiliation(s)
- Fang Wu
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Hangping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Institute of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Fenping Zheng
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Shengjie Tang
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Xihua Lin
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Lin Li
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Jiaqiang Zhou
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Hong Li
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
15
|
Sun PF, Tian T, Chen LN, Fu RG, Xu SS, Ai H, Wang B, Zhang J, Si RY, Chai Z, Cooper ME, Ren ST. Ultrasound Combined with Microbubbles Enhances the Effects of Methylprednisolone in Lipopolysaccharide-Induced Human Mesangial Cells. J Pharmacol Exp Ther 2018; 365:476-484. [PMID: 29549156 DOI: 10.1124/jpet.117.246223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/09/2018] [Indexed: 03/08/2025] Open
Abstract
A novel drug delivery system mediated by ultrasound (US) combined with microbubbles (MBs) (US+MB) could improve local drug concentration to enhance its efficacy. To investigate the influence of US+MB on methylprednisolone (MP), the effect of US+MB combined with MP (US+MB+MP) on lipopolysaccharide (LPS)-induced human mesangial cells (HMCs) and the underlying mechanism were explored in this study. The results revealed that HMCs treated with LPS underwent significant proliferation and exhibited an increase in nuclear transcription factor-κB (NF-κB) and transforming growth factor-β1 (TGF-β1) expression and a decrease in cellular apoptosis. This effect was significantly inhibited by MP (30-100 μg/ml), US combined with MBs (3.22 × 107 and 8.05 × 107 bubbles/ml), and US combined with both MBs (1.29 × 107 bubbles/ml) and MP (12 μg/ml) (US+MB1+MP12). The effect of US+MB1+MP12 was better than the effect of 12 μg/ml of MP alone and was similar to the effect of 100 μg/ml of MP. Additionally, the intracellular free MP content was significantly higher in the US+MB1+MP12 group than in the MP12 group. US combined with MBs not only inhibited LPS-induced HMC proliferation and NF-κB and TGF-β1 expression and increased cellular apoptosis but also synergized with the pharmacologic effect of MP. The mechanism is partially due to the US-assisted MB local drug delivery and the anti-inflammatory effect induced by US combined with MBs.
Collapse
Affiliation(s)
- Peng-Fei Sun
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Tian Tian
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Li-Na Chen
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Rong-Guo Fu
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Shan-Shan Xu
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Hong Ai
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Bing Wang
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Jian Zhang
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Ruo-Yan Si
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Zhonglin Chai
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Mark E Cooper
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| | - Shu-Ting Ren
- Department of Pathology (P.-F.S., T.T., B.W., J.Z., S.-T.R.), Department of Pharmacology (L.-N.C.), and Therapeutic Vaccines Engineering Center of Shaanxi Province (B.W.), School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China; Department of Pathology, Xi'an City Center Hospital, Xi'an, People's Republic of China (P.-F.S.); Department of Nephrology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (R.-G.F.); Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China (S.-S.X.); Department of Ultrasound, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China (H.A.); Shanghai Medical College, Fudan University, Shanghai, People's Republic of China (R.-Y.S.); and Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia (Z.C., M.E.C.)
| |
Collapse
|
16
|
Abstract
This study was aimed to clarify the effect of honokiol (Hon) on the activity of Cytochrome P450 (CYP450) enzymes, and the level of mRNA expression of liver and kidney transporters in type 2 diabetic rats induced by high-fat diet and strepotozotocin. Rats were randomly divided into normal control (NC) group, diabetic control (DC) group and Hon groups (n = 6). The activities of hepatic CYP1A2, CYP2E1, CYP2C, CYP2B, CYP3A and CYP4A, and the mRNA expression levels of hepatic and renal transporters, were determined. Compared to the NC group, the activities of CYP1A2, CYP2E1, CYP4A and CYP2C in DC group were increased by 2.36-, 2.10-, 2.55- and 1.86-fold, respectively. The mRNA expression levels of hepatic Oat2, Oatp2b1 and Oatp1a5, and renal Oct1, Octn2, Oatp2b1 and Oatp1a5, were significantly down-regulated, while the mRNA expression levels of hepatic Octn2, Oatp3a1, Oatp1a1 and Mdr2, and renal Oat2, Mrp4 and Bcrp, were significantly upregulated. Compared to the DC group, Hon treatment significantly inhibited the activity of hepatic CYP2E1, CYP4A, 3A and CYP1A2 by 45.6%, 29.2%, 22.7% and 20.7% in Hon high dose group, respectively. Moreover, Hon treatment significantly inhibited the mRNA expression levels of renal Bcrp and Mrp4 by 2.63-fold and 1.54-fold, while significantly upregulated the mRNA expression levels of hepatic Oat2 and Oatp2b1 by 1.52-fold and 1.54-fold in Hon high dose group, respectively. The results suggested that under the diabetes condition, the changes of CYP450 activity and transporter expression inevitably interfere the normal transport, metabolism and efficacy of drugs. The present work firstly reported that Hon treatment ameliorated the abnormal change of hepatic CYP activity (including CYP2E1, CYP4A and CYP1A2) and the transporter mRNA expression (including hepatic Oat2 and Oatp2b1, renal Bcrp and Mrp4) in type 2 diabetic rats induced by high-fat diet and strepotozotocin, which are associated with the occurrence and development of diabetes.
Collapse
|
17
|
Bhatt NP, Park JY, Lee HJ, Kim SS, Kwon YS, Chun W. Apocynin protects mesangial cells from lipopolysaccharide-induced inflammation by exerting heme oxygenase 1-mediated monocyte chemoattractant protein-1 suppression. Int J Mol Med 2017; 40:1294-1301. [DOI: 10.3892/ijmm.2017.3090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
|
18
|
Vergani E, Di Guardo L, Dugo M, Rigoletto S, Tragni G, Ruggeri R, Perrone F, Tamborini E, Gloghini A, Arienti F, Vergani B, Deho P, De Cecco L, Vallacchi V, Frati P, Shahaj E, Villa A, Santinami M, De Braud F, Rivoltini L, Rodolfo M. Overcoming melanoma resistance to vemurafenib by targeting CCL2-induced miR-34a, miR-100 and miR-125b. Oncotarget 2016; 7:4428-41. [PMID: 26684239 PMCID: PMC4826216 DOI: 10.18632/oncotarget.6599] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/25/2015] [Indexed: 01/06/2023] Open
Abstract
In melanoma, the adaptative cell response to BRAF inhibitors includes altered patterns of cytokine production contributing to tumor progression and drug resistance. Among the factors produced by PLX4032-resistant melanoma cell lines, CCL2 was higher compared to the sensitive parental cell lines and increased upon drug treatment. CCL2 acted as an autocrine growth factor for melanoma cells, stimulating the proliferation and resistance to apoptosis. In patients, CCL2 is detected in melanoma cells in tumors and in plasma at levels that correlate with tumor burden and lactate dehydrogenase. Vemurafenib treatment increased the CCL2 levels in plasma, whereas the long-term clinical response was associated with low CCL2 levels.Increased CCL2 production was associated with miRNA deregulation in the resistant cells. miR-34a, miR-100 and miR-125b showed high expression in both resistant cells and in tumor biopsies that were obtained from treated patients, and they were involved in the control of cell proliferation and apoptosis. Inhibition of CCL2 and of the selected miRNAs restored both the cell apoptosis and the drug efficacy in resistant melanoma cells. Therefore, CCL2 and miRNAs are potential prognostic factors and attractive targets for counteracting treatment resistance in metastatic melanoma.
Collapse
Affiliation(s)
- Elisabetta Vergani
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lorenza Di Guardo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Dugo
- Functional Genomics and Bioinformatics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Rigoletto
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gabrina Tragni
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberta Ruggeri
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Tamborini
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Flavio Arienti
- Immunohematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Vergani
- Consorzio MIA, Microscopy and Image Analysis, University of Milan Bicocca, Monza, Italy
| | - Paola Deho
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Functional Genomics and Bioinformatics Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Viviana Vallacchi
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paola Frati
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eriomina Shahaj
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonello Villa
- Consorzio MIA, Microscopy and Image Analysis, University of Milan Bicocca, Monza, Italy
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo De Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Licia Rivoltini
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Rodolfo
- Immunotherapy Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
19
|
The neutrophil elastase inhibitor, sivelestat, attenuates sepsis-related kidney injury in rats. Int J Mol Med 2016; 38:767-75. [PMID: 27430552 PMCID: PMC4990314 DOI: 10.3892/ijmm.2016.2665] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/24/2016] [Indexed: 12/18/2022] Open
Abstract
Sepsis-induced acute kidney injury (AKI) represents a major cause of mortality in intensive care units. Sivelestat, a selective inhibitor of neutrophil elastase (NE), can attenuate sepsis-related acute lung injury. However, whether sivelestat can preserve kidney function during sepsis remains unclear. In this study, we thus examined the effects of sivelestat on sepsis-related AKI. Cecal ligation and puncture (CLP) was performed to induce multiple bacterial infection in male Sprague-Dawley rats, and subsequently, 50 or 100 mg/kg sivelestat were administered by intraperitoneal injection immediately after the surgical procedure. In the untreated rats with sepsis, the mean arterial pressure (MAP) and glomerular filtration rate (GFR) were decreased, whereas serum blood urea nitrogen (BUN) and neutrophil gelatinase-associated lipocalin (NGAL) levels were increased. We found that sivelestat promoted the survival of the rats with sepsis, restored the impairment of MAP and GFR, and inhibited the increased BUN and NGAL levels; specifically, the higher dose was more effective. In addition, sivelestat suppressed the CLP-induced macrophage infiltration, the overproduction of pro-inflammatory mediators (tumor necrosis factor-α, interleukin-1β, high-mobility group box 1 and inducible nitric oxide synthase) and serine/threonine kinase (Akt) pathway activation in the rats. Collectively, our data suggest that the inhibition of NE activity with the inhibitor, sivelestat, is beneficial in ameliorating sepsis-related kidney injury.
Collapse
|
20
|
Chuang DY, Simonyi A, Cui J, Lubahn DB, Gu Z, Sun GY. Botanical Polyphenols Mitigate Microglial Activation and Microglia-Induced Neurotoxicity: Role of Cytosolic Phospholipase A2. Neuromolecular Med 2016; 18:415-25. [PMID: 27339657 DOI: 10.1007/s12017-016-8419-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/11/2016] [Indexed: 01/17/2023]
Abstract
Microglia play a significant role in the generation and propagation of oxidative/nitrosative stress, and are the basis of neuroinflammatory responses in the central nervous system. Upon stimulation by endotoxins such as lipopolysaccharides (LPS), these cells release pro-inflammatory factors which can exert harmful effects on surrounding neurons, leading to secondary neuronal damage and cell death. Our previous studies demonstrated the effects of botanical polyphenols to mitigate inflammatory responses induced by LPS, and highlighted an important role for cytosolic phospholipase A2 (cPLA2) upstream of the pro-inflammatory pathways (Chuang et al. in J Neuroinflammation 12(1):199, 2015. doi: 10.1186/s12974-015-0419-0 ). In this study, we investigate the action of botanical compounds and assess whether suppression of cPLA2 in microglia is involved in the neurotoxic effects on neurons. Differentiated SH-SY5Y neuroblastoma cells were used to test the neurotoxicity of conditioned medium from stimulated microglial cells, and WST-1 assay was used to assess for the cell viability of SH-SY5Y cells. Botanicals such as quercetin and honokiol (but not cyanidin-3-O-glucoside, 3CG) were effective in inhibiting LPS-induced nitric oxide (NO) production and phosphorylation of cPLA2. Conditioned medium from BV-2 cells stimulated with LPS or IFNγ caused neurotoxicity to SH-SY5Y cells. Decrease in cell viability could be ameliorated by pharmacological inhibitors for cPLA2 as well as by down-regulating cPLA2 with siRNA. Botanicals effective in inhibition of LPS-induced NO and cPLA2 phosphorylation were also effective in ameliorating microglial-induced neurotoxicity. Results demonstrated cytotoxic factors from activated microglial cells to cause damaging effects to neurons and potential use of botanical polyphenols to ameliorate the neurotoxic effects.
Collapse
Affiliation(s)
- Dennis Y Chuang
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA.,Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA.,Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA
| | - Agnes Simonyi
- Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA.,Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Jiankun Cui
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA.,Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA.,Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Dennis B Lubahn
- Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA.,Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Zezong Gu
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA.,Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA.,Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA.,Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Grace Y Sun
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA. .,Center for Translational Neuroscience, University of Missouri, Columbia, MO, USA. .,Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA. .,Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA.
| |
Collapse
|
21
|
Alpha-lipoic acid exerts anti-inflammatory effects on lipopolysaccharide-stimulated rat mesangial cells via inhibition of nuclear factor kappa B (NF-κB) signaling pathway. Inflammation 2015; 38:510-9. [PMID: 24962643 DOI: 10.1007/s10753-014-9957-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sepsis is often initiated by invasive infection, characterized by overwhelming induction of pro-inflammatory cytokines. The incidence and mortality of sepsis and the associated development of acute kidney injury (AKI) remain high, and lines of research into potential treatments are needed. This study was conducted to investigate effects of alpha-lipoic acid (ALA) on septic AKI in vitro. ALA of 200 or 400 μM was used to pretreat rat HBZY-1 mesangial cells before commencement of 1 μg/mL lipopolysaccharide (LPS). Our data indicated that ALA pretreatment reduced LPS-stimulated release of inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1 beta (IL-1β), as well as IL-6, in HBZY-1 cell supernatant. Moreover, LPS-induced expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) was inhibited by ALA pretreatment, and consequently, the secretion levels of their respective enzymatic products prostaglandin E2 (PGE2) and nitric oxide (NO) were significantly decreased. LPS-enhanced phosphorylation of nuclear factor kappa B (NF-κB) inhibitor alpha (IκBα) and IκB kinase alpha/beta (IKKα/β) and nuclear translocation of NF-κB subunit p65 in HBZY-1 cells were inhibited by ALA pretreatment. Additionally, the NF-κB inhibitor N-acetylcysteine (NAC) exerted similar inhibitory effects as ALA on COX-2 and iNOS expression. In summary, our study demonstrates that ALA mitigates LPS-induced inflammatory responses in rat mesangial cells probably via inhibition of NF-κB signaling pathway, suggesting a therapeutic potential of ALA in AKI related to sepsis.
Collapse
|
22
|
You Y, Qin Y, Lin X, Yang F, Li J, Sooranna SR, Pinhu L. Methylprednisolone attenuates lipopolysaccharide-induced Fractalkine expression in kidney of Lupus-prone MRL/lpr mice through the NF-kappaB pathway. BMC Nephrol 2015; 16:148. [PMID: 26310926 PMCID: PMC4551515 DOI: 10.1186/s12882-015-0145-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 03/05/2015] [Accepted: 08/20/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Fractalkine (FKN) is involved in the occurrence and development of human lupus nephritis. It is known to be upregulated by lipopolysaccharide (LPS) as a stimulus in vivo. MRL/lpr mice have been used as an in vivo model to study lupus nephritis. Methylprednisolone (MP) is used widely in the clinical treatment of progressive glomerular diseases such as lupus nephritis. The aim of this study is to explore the mechanism of LPS induced FKN expression and to determine whether other molecular mechanisms contribute to the signaling pathway of MP action in MRL/lpr mice. METHODS Forty-eight female MRL/lpr mice at 12 weeks of age were randomly distributed into six groups. Each group received various treatments for 8 weeks by receiving twice weekly intraperitoneal injections of (1) MP (MP-treated mice), of (2) SC-514 (SC-514-induced mice), of (3) normal saline and a single injection of LPS (LPS-induced mice), of (4) MP and a single injection of LPS (LPS + MP mice), of (5) SC-514 and a single injection of LPS (LPS + SC mice) and of (6) normal saline (control mice). One-way ANOVA was used for data analysis and P value <0.05 was considered statistically significantly. RESULTS The expression of FKN and NF-kappaB p65 mRNA was detected by qPCR. The expression of FKN protein and the activation of NF-kappaB p65 were detected by immunohistochemistry and western blots respectively. The expression of FKN in the kidney of LPS induced mice was significantly increased and this was mediated by increased expression of NF-κB p65 and an increase in NF-kappaB phospho-p65. MP reduced proteinuria and ameliorated the renal damage in MRL/lpr mice. MP as well as the NF-kappaB inhibitor, SC-514, inhibited the LPS-induced increase of expression of FKN and the activation of NF-kappaB. CONCLUSIONS The results indicate that MP attenuates LPS-induced FKN expression in kidney of MRL/lpr mice through the NF-kappaB pathway.
Collapse
Affiliation(s)
- Yanwu You
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| | - Yueqiu Qin
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| | - Xu Lin
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| | - Fafen Yang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| | - Jun Li
- Department of Intensive Care Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| | - Suren R Sooranna
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.
| | - Liao Pinhu
- Department of Intensive Care Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
23
|
Li S, Gao X, Wu X, Wu Z, Cheng L, Zhu L, Shen D, Tong X. Parthenolide inhibits LPS-induced inflammatory cytokines through the toll-like receptor 4 signal pathway in THP-1 cells. Acta Biochim Biophys Sin (Shanghai) 2015; 47:368-375. [PMID: 25841439 DOI: 10.1093/abbs/gmv019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/20/2015] [Indexed: 02/07/2023] Open
Abstract
Parthenolide (PTL) shows potent anti-inflammatory and anti-cancer activities. In the present study, the molecular mechanisms of PTL's activities were explored in lipopolysaccharide (LPS)-induced human leukemia monocytic THP-1 cells and human primary monocytes. The 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium salt (MTS) assay was used to analyze the effect of PTL on THP-1 cell viability. Enzyme-linked immunosorbent assay was used to determine the effect of PTL on LPS-induced inflammatory cytokine secretion. Flow cytometry and quantitative real-time polymerase chain reaction were used to assess the effect of PTL on LPS-induced toll-like receptor 4 (TLR4) expression. Phosphorylation levels of signaling molecules were determined by western blot analysis. Results showed that PTL <12.5 μM did not significantly affect THP-1 cells viability. LPS treatment led to a marked up-regulation of interleukin (IL)-6, IL-1β, IL-8, IL-12p40, tumor necrosis factor-α, IL-18, and NO in THP-1 cells. However, PTL inhibited the expression of these cytokines in a dose-dependent manner, with IC50 values of 1.091-2.620 μM. PTL blocked TLR4 expression with an IC50 value of 1.373 μM as determined by the flow cytometry analysis, and this blocking effect was verified at both protein and mRNA levels. Up-regulation of phosphorylation levels of extracellular signal-regulated kinase 1/2, Jun N-terminal kinase, p38, nuclear factor κB (NF-κB) p65, and IκBα and up-regulation of expressions of other molecules (inducible nitric oxide synthase, TLR4, and TNF receptor-associated factor 6) induced by LPS were abolished by PTL in a dose-dependent manner. The anti-inflammatory mechanisms of PTL operate partly through the TLR4-mediated mitogen-activated protein kinase and NF-κB signaling pathways. Therefore, TLR4 may be a new target for anti-inflammation therapies.
Collapse
Affiliation(s)
- Shuangshuang Li
- Department of Hematology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China Department of Hematology, Zhejiang Provincial People's Hospital, Hangzhou 310003, China
| | - Xiangli Gao
- Department of Hematology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Zhigang Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Linfang Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Lifen Zhu
- Department of Hematology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Dan Shen
- Department of Hematology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China
| | - Xiangmin Tong
- Department of Hematology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China Department of Hematology, Zhejiang Provincial People's Hospital, Hangzhou 310003, China
| |
Collapse
|
24
|
Effects of honokiol on sepsis-induced acute kidney injury in an experimental model of sepsis in rats. Inflammation 2015; 37:1191-9. [PMID: 24531855 DOI: 10.1007/s10753-014-9845-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) is a severe complication of sepsis, which largely contributes to the high mortality rate of sepsis. Honokiol, a natural product isolated from Magnolia officinalis (Houpo), has been shown to exhibit anti-inflammatory and antioxidant properties. Here, we investigated the effects of honokiol on sepsis-associated AKI in rats subjected to cecal ligation and puncture (CLP). We found that the administration of honokiol improved the survival of septic rats. Periodic acid-Schiff stain revealed that the morphological changes of kidney tissues in CLP rats were restored after honokiol treatment. Furthermore, honokiol reduced CLP-induced oxidative stress and inflammatory cytokine production. The levels of nitric oxide (NO) and inducible NO synthetase (iNOS) were attenuated by honokiol in septic rats. Finally, honokiol inhibited CLP-induced activation of NF-κB signaling in CLP rats. Our findings suggest that honokiol might be used as a potential therapeutic agent for complications of sepsis, especially for sepsis-induced AKI.
Collapse
|
25
|
Wang JJ, Zhao R, Liang JC, Chen Y. Antidiabetic and Anti-oxidative Effects of Honokiol on Diabetic Rats Induced by High-fat Diet and Streptozotocin. CHINESE HERBAL MEDICINES 2014. [DOI: 10.1016/s1674-6384(14)60005-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
26
|
Chuang DY, Chan MH, Zong Y, Sheng W, He Y, Jiang JH, Simonyi A, Gu Z, Fritsche KL, Cui J, Lee JC, Folk WR, Lubahn DB, Sun AY, Sun GY. Magnolia polyphenols attenuate oxidative and inflammatory responses in neurons and microglial cells. J Neuroinflammation 2013; 10:15. [PMID: 23356518 PMCID: PMC3576246 DOI: 10.1186/1742-2094-10-15] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 01/17/2013] [Indexed: 11/29/2022] Open
Abstract
Background The bark of magnolia has been used in Oriental medicine to treat a variety of remedies, including some neurological disorders. Magnolol (Mag) and honokiol (Hon) are isomers of polyphenolic compounds from the bark of Magnolia officinalis, and have been identified as major active components exhibiting anti-oxidative, anti-inflammatory, and neuroprotective effects. In this study, we investigate the ability of these isomers to suppress oxidative stress in neurons stimulated by the ionotropic glutamate receptor agonist N-methyl-D-aspartate (NMDA) and oxidative and inflammatory responses in microglial cells activated by interferon-γ (IFNγ) and lipopolysaccharide (LPS). We also attempt to elucidate the mechanism and signaling pathways involved in cytokine-induced production of reactive oxygen species (ROS) in microglial cells. Methods Dihydroethidium (DHE) was used to assay superoxide production in neurons, while CM-H2DCF-DA was used to test for ROS production in murine (BV-2) and rat (HAPI) immortalized microglial cells. NADPH oxidase inhibitors (for example, diphenyleneiodonium (DPI), AEBSF, and apocynin) and immunocytochemistry targeting p47phox and gp91phox were used to assess the involvement of NADPH oxidase. Western blotting was used to assess iNOS and ERK1/2 expression, and the Griess reaction protocol was employed to determine nitric oxide (NO) concentration. Results Exposure of Hon and Mag (1–10 μM) to neurons for 24 h did not alter neuronal viability, but both compounds (10 μM) inhibited NMDA-stimulated superoxide production, a pathway known to involve NADPH oxidase. In microglial cells, Hon and Mag inhibited IFNγ±LPS-induced iNOS expression, NO, and ROS production. Studies with inhibitors and immunocytochemical assay further demonstrated the important role of IFNγ activating the NADPH oxidase through the p-ERK-dependent pathway. Hon and, to a lesser extent, Mag inhibited IFNγ-induced p-ERK1/2 and its downstream pathway for ROS and NO production. Conclusion This study highlights the important role of NADPH oxidase in mediating oxidative stress in neurons and microglial cells and has unveiled the role of IFNγ in stimulating the MAPK/ERK1/2 signaling pathway for activation of NADPH oxidase in microglial cells. Hon and Mag offer anti-oxidative or anti-inflammatory effects, at least in part, through suppressing IFNγ-induced p-ERK1/2 and its downstream pathway.
Collapse
Affiliation(s)
- Dennis Y Chuang
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhang P, Liu X, Zhu Y, Chen S, Zhou D, Wang Y. Honokiol inhibits the inflammatory reaction during cerebral ischemia reperfusion by suppressing NF-κB activation and cytokine production of glial cells. Neurosci Lett 2012; 534:123-7. [PMID: 23262090 DOI: 10.1016/j.neulet.2012.11.052] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 11/14/2012] [Accepted: 11/28/2012] [Indexed: 12/16/2022]
Abstract
This study was designed to investigate the effects of honokiol, a neuroprotective agent, on cerebral edema in cerebral ischemia reperfusion (IR) mice and its mechanism of anti-inflammation. Honokiol (0.7-70μg/kg) significantly reduced brain water contents and decreased the exudation of Evans blue dye from brain capillaries in cerebral IR mice. Honokiol (0.1-10μM) significantly reduced the p65 subunit level of NF-κB in the nucleus of primary culture-microglia. It (0.01-10μM) evidently reduced nitric oxide (NO) level in the microglia culture medium and in the microglia and astrocytes coculture medium. Honokiol (0.01-10μM) significantly decreased the level of TNF-α in the microglia medium or coculture cell medium. Honokiol (10μM) decreased the level of Regulated upon Activation Normal T-cell Expressed and Secreted (RANTES/CCL5) protein in medium of microglia or astrocytes. In conclusion, Honokiol has a potent anti-inflammatory effect in cerebral ischemia-reperfusion mice and this effect might be attributed to its inhibition ability on the NF-κB activation, consequently blocking the production of inflammatory factors including: NO, tumor necrosis factor-α (TNF-α) and RANTES/CCL5 in glial cells. These results provide evidence for the anti-inflammatory effect of honokiol for the potential treatment of ischemic stroke.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | | | | | | | | | | |
Collapse
|
28
|
Park J, Song KH, Ha H. Lipopolysaccharide increases monocyte binding to mesangial cells through fractalkine and its receptor. Transplant Proc 2012; 44:1029-31. [PMID: 22564617 DOI: 10.1016/j.transproceed.2012.03.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Fractalkine (CX3CL1) is a unique chemokine that functions not only as a chemokine but also as an adhesion molecule. Fractalkine plays an important role in the recruitment of macrophages into the kidneys by binding to its specific receptor CX3CR1, and renal fractalkine expression was shown to be increased in chronic renal allograft rejection. Considering that microcapillary inflammation is a key feature of chronic renal allograft rejection, the present study examined whether monocytes bind to mesangial cells cultured in the presence of lipopolysaccharide (LPS) through fractalkine/CX3CR1 in order to understand their regulation with respect to inflammation-induced renal allograft dysfunction. Mouse mesangial cells were stimulated with LPS in the presence or absence of fractalkine or CX3CR1 siRNA. Calcein-AM-labeled monocytes were used to evaluate monocyte binding. Fractalkine and CX3CR1 mRNA and protein expression were measured by real-time quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. LPS at 100 ng/mL significantly increased monocyte binding to mesangial cells. Each siRNA against fractalkine or CX3CR1 effectively inhibited LPS-induced monocyte-mesangial cell binding. Fractalkine and CX3CR1 mRNA expression were enhanced in mesangial cells stimulated with LPS. Fractalkine protein synthesis in media and lysate of mesangial cells were also induced by LPS. These results demonstrated that LPS induces monocyte-mesangial cell binding through the fractalkine/CX3CR1 system and suggested that fractalkine/CX3CR1 system may contribute to renal inflammation leading to chronic renal allograft rejection.
Collapse
Affiliation(s)
- J Park
- Department of Bioinspired Science and Division of Life and Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | | | | |
Collapse
|
29
|
Lin JM, Prakasha Gowda AS, Sharma AK, Amin S. In vitro growth inhibition of human cancer cells by novel honokiol analogs. Bioorg Med Chem 2012; 20:3202-11. [PMID: 22533983 DOI: 10.1016/j.bmc.2012.03.062] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 03/19/2012] [Accepted: 03/27/2012] [Indexed: 11/26/2022]
Abstract
Honokiol possesses many pharmacological activities including anti-cancer properties. Here in, we designed and synthesized honokiol analogs that block major honokiol metabolic pathway which may enhance their effectiveness. We studied their cytotoxicity in human cancer cells and evaluated possible mechanism of cell cycle arrest. Two analogs, namely 2 and 4, showed much higher growth inhibitory activity in A549 human lung cancer cells and significant increase of cell population in the G0-G1 phase. Further elucidation of the inhibition mechanism on cell cycle showed that analogs 2 and 4 inhibit both CDK1 and cyclin B1 protien levels in A549 cells.
Collapse
Affiliation(s)
- Jyh Ming Lin
- Department of Biochemistry & Molecular Biology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA.
| | | | | | | |
Collapse
|
30
|
Kaushik DK, Mukhopadhyay R, Kumawat KL, Gupta M, Basu A. Therapeutic targeting of Krüppel-like factor 4 abrogates microglial activation. J Neuroinflammation 2012; 9:57. [PMID: 22429472 PMCID: PMC3325890 DOI: 10.1186/1742-2094-9-57] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/19/2012] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Neuroinflammation occurs as a result of microglial activation in response to invading micro-organisms or other inflammatory stimuli within the central nervous system. According to our earlier findings, Krüppel-like factor 4 (Klf4), a zinc finger transcription factor, is involved in microglial activation and subsequent release of proinflammatory cytokines, tumor necrosis factor alpha, macrophage chemoattractant protein-1 and interleukin-6 as well as proinflammatory enzymes, inducible nitric oxide synthase and cyclooxygenase-2 in lipopolysaccharide-treated microglial cells. Our current study focuses on finding the molecular mechanism of the anti-inflammatory activities of honokiol in lipopolysaccharide-treated microglia with emphasis on the regulation of Klf4. METHODS For in vitro studies, mouse microglial BV-2 cell lines as well as primary microglia were treated with 500 ng/mL lipopolysaccharide as well as 1 μM and 10 μM of honokiol. We cloned full-length Klf4 cDNA in pcDNA3.1 expression vector and transfected BV-2 cells with this construct using lipofectamine for overexpression studies. For in vivo studies, brain tissues were isolated from BALB/c mice treated with 5 mg/kg body weight of lipopolysaccharide either with or without 2.5 or 5 mg/kg body weight of honokiol. Expression of Klf4, cyclooxygenase-2, inducible nitric oxide synthase and phospho-nuclear factor-kappa B was measured using immunoblotting. We also measured the levels of cytokines, reactive oxygen species and nitric oxide in different conditions. RESULTS Our findings suggest that honokiol can substantially downregulate the production of proinflammatory cytokines and inflammatory enzymes in lipopolysaccharide-stimulated microglia. In addition, honokiol downregulates lipopolysaccharide-induced upregulation of both Klf4 and phospho-nuclear factor-kappa B in these cells. We also found that overexpression of Klf4 in BV-2 cells suppresses the anti-inflammatory action of honokiol. CONCLUSIONS Honokiol potentially reduces inflammation in activated microglia in a Klf4-dependent manner.
Collapse
|