1
|
Patel B, Patel S, Modi F, Patel A, Gelat B, Tanavde V, Vasavada A, Johar K. Combination of paclitaxel with rosiglitazone induces synergistic cytotoxic effects in ovarian cancer cells. Sci Rep 2024; 14:30672. [PMID: 39730440 DOI: 10.1038/s41598-024-74277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/24/2024] [Indexed: 12/29/2024] Open
Abstract
Ovarian cancer is known to be a challenging disease to detect at an early stage and is a major cause of death among women. The current treatment for ovarian cancer typically involves a combination of surgery and the use of drugs such as platinum-based cytotoxic agents, anti-angiogenic drugs, etc. However, current treatment methods are not always effective in preventing the recurrence of ovarian cancer. As a result, the treatments administered after a relapse need to be more aggressive, leading to increased toxicity and drug resistance. To address this issue, researchers are exploring the potential of combining existing anticancer agents with novel or repurposed drugs to reduce the side effects and improve the effectiveness of treatment. In this study, we have investigated the use of rosiglitazone, a well-known anti-diabetic drug, in combination with the chemotherapeutic drug, paclitaxel for the prevention of ovarian cancer. The study utilized the SKOV-3 ovarian cancer cell line to assess the effects of this combination treatment. The results of the study showed that the combination of paclitaxel with rosiglitazone inhibited cell proliferation at much lower concentrations of paclitaxel as compared to paclitaxel alone. The combined treatment also induced cell cycle arrest at the G2/M phase and increased apoptosis by altering the mitochondrial membrane potential of the cells. Additionally, the combination treatment activated the PPAR-γ pathway and downregulated expression of genes associated with cancer stemness, such as NANOG, OCT4, and EHF. Furthermore, the CAM assay substantiated the anti-angiogenic potential of the synergistic treatment of paclitaxel and rosiglitazone. The findings of the study suggest that repurposing rosiglitazone as an anticancer agent in combination with paclitaxel has immense potential to target cancer cell cycle progression and apoptosis, making it a promising therapeutic approach for sensitizing chemo-resistant population of ovarian cancer cells.
Collapse
Affiliation(s)
- Binita Patel
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Shanaya Patel
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Foram Modi
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Aditi Patel
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Brijesh Gelat
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Vivek Tanavde
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Ahmedabad, 380009, Gujarat, India
| | - Abhay Vasavada
- Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, 380052, Gujarat, India
| | - Kaid Johar
- Department of Zoology, Biomedical Technology, Human Genetics, and WBC, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India.
| |
Collapse
|
2
|
Lee IT, Yang CC, Yang CM. Harnessing peroxisome proliferator-activated receptor γ agonists to induce Heme Oxygenase-1: a promising approach for pulmonary inflammatory disorders. Cell Commun Signal 2024; 22:125. [PMID: 38360670 PMCID: PMC10868008 DOI: 10.1186/s12964-024-01501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024] Open
Abstract
The activation of peroxisome proliferator-activated receptor (PPAR)-γ has been extensively shown to attenuate inflammatory responses in conditions such as asthma, acute lung injury, and acute respiratory distress syndrome, as demonstrated in animal studies. However, the precise molecular mechanisms underlying these inhibitory effects remain largely unknown. The upregulation of heme oxygenase-1 (HO-1) has been shown to confer protective effects, including antioxidant, antiapoptotic, and immunomodulatory effects in vitro and in vivo. PPARγ is highly expressed not only in adipose tissues but also in various other tissues, including the pulmonary system. Thiazolidinediones (TZDs) are highly selective agonists for PPARγ and are used as antihyperglycemic medications. These observations suggest that PPARγ agonists could modulate metabolism and inflammation. Several studies have indicated that PPARγ agonists may serve as potential therapeutic candidates in inflammation-related diseases by upregulating HO-1, which in turn modulates inflammatory responses. In the respiratory system, exposure to external insults triggers the expression of inflammatory molecules, such as cytokines, chemokines, adhesion molecules, matrix metalloproteinases, and reactive oxygen species, leading to the development of pulmonary inflammatory diseases. Previous studies have demonstrated that the upregulation of HO-1 protects tissues and cells from external insults, indicating that the induction of HO-1 by PPARγ agonists could exert protective effects by inhibiting inflammatory signaling pathways and attenuating the development of pulmonary inflammatory diseases. However, the mechanisms underlying TZD-induced HO-1 expression are not well understood. This review aimed to elucidate the molecular mechanisms through which PPARγ agonists induce the expression of HO-1 and explore how they protect against inflammatory and oxidative responses.
Collapse
Affiliation(s)
- I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, 333008, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, 242062, Taiwan.
| |
Collapse
|
3
|
Karmakar E, Das N, Mukherjee B, Das P, Mukhopadhyay S, Roy SS. Lipid-induced alteration in retinoic acid signaling leads to mitochondrial dysfunction in HepG2 and Huh7 cells. Biochem Cell Biol 2023. [PMID: 36787544 DOI: 10.1139/bcb-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
A surfeit of mitochondrial reactive oxygen species (ROS) and inflammation serve as obligatory mediators of lipid-associated hepatocellular maladies. While retinoid homeostasis is essential in restoring systemic energy balance, its role in hepatic mitochondrial function remains elusive. The role of lecithin-retinol acyltransferase (LRAT) in maintenance of retinoid homeostasis is appreciated earlier; however, its role in modulating retinoic acid (RA) bioavailability upon lipid-imposition is unexplored. We identified LRAT overexpression in high-fat diet (HFD)-fed rats and palmitate-treated hepatoma cells. Elevation in LRAT expression depletes RA production and deregulates RA signaling. This altered RA metabolism enhances fat accumulation, accompanied by inflammation that leads to impaired mitochondrial function through enhanced ROS generation. Hence, LRAT inhibition could be a novel approach preventing lipid-induced mitochondrial dysfunction in hepatoma cells.
Collapse
Affiliation(s)
- Eshani Karmakar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Nabanita Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Bijnor-sisendi Road, Lucknow, Uttar Pradesh, 226002, India
| | - Bidisha Mukherjee
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata, 700020, India
| | - Prosenjit Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Satinath Mukhopadhyay
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, 244, A.J.C. Bose Road, Kolkata, 700020, India
| | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, India.,Academy of Scientific & Innovative Research (AcSIR), India
| |
Collapse
|
4
|
Wang Z, Shen W, Li X, Feng Y, Qian K, Wang G, Gao Y, Xu X, Zhang S, Yue L, Cao J. The PPARγ Agonist Rosiglitazone Enhances the Radiosensitivity of Human Pancreatic Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3099-3110. [PMID: 32801648 PMCID: PMC7410396 DOI: 10.2147/dddt.s242557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/03/2020] [Indexed: 01/29/2023]
Abstract
Purpose As radiation therapy is widely used for the management of pancreatic cancer, identifying novel targets to improve the radiosensitivity of cancer cells is beneficial. Rosiglitazone, a specific peroxisome proliferator-activated receptor γ (PPARγ) agonist, has an inhibitory effect on various types of cancer cells. The purpose of this paper is to investigate the effect of rosiglitazone on the radiosensitivity of pancreatic cancer cells and the potential mechanism. Materials and Methods PPARγ expression in pancreatic cancer and adjacent tissues was evaluated using immunohistochemistry analysis. The viability, migration and invasion ability of PANC1 and PaTu8988 cells were detected using MTT assay, scratch-wound assay and transwell invasion assay. The effect of rosiglitazone on radiosensitivity of the cells was determined using the clonogenic assay. PANC1 cells were inoculated into BALB/c mice to establish tumors. Microarray was used to investigate changes of genes involved. Results Higher PPARγ expression was demonstrated in pancreatic cancer tissues compared with para-carcinoma tissues. Rosiglitazone inhibited the cell viability and enhanced the radiation-induced anti-migration and anti-invasion effect. Rosiglitazone potentiated the radiosensitivity of pancreatic cancer cells and PANC1 xenografts. Microarray analysis revealed that rosiglitazone plus radiation altered the expression of multiple genes and affected multiple pathways. Conclusion Rosiglitazone enhances the radiosensitivity of human pancreatic cancer cells in vitro and in vivo via complex mechanisms.
Collapse
Affiliation(s)
- Zhenyu Wang
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Wenhao Shen
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Xugang Li
- Department of Radiotherapy, Anshan Cancer Hospital, Anshan 114036, People's Republic of China
| | - Yang Feng
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Kun Qian
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Gaoren Wang
- Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226361, People's Republic of China
| | - Yiying Gao
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Xiaohui Xu
- Department of General Surgery, The First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou 215400, People's Republic of China
| | - Shuyu Zhang
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Ling Yue
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| | - Jianping Cao
- School of Radiation Medicine and Protection (SRMP) of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Suzhou 215123, People's Republic of China
| |
Collapse
|
5
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
6
|
Biondo LA, Teixeira AAS, de O. S. Ferreira KC, Neto JCR. Pharmacological Strategies for Insulin Sensitivity in Obesity and Cancer: Thiazolidinediones and Metformin. Curr Pharm Des 2020; 26:932-945. [PMID: 31969093 DOI: 10.2174/1381612826666200122124116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/21/2019] [Indexed: 12/19/2022]
Abstract
Background:
Chronic diseases, such as obesity and cancer, have high prevalence rates. Both diseases
have hyperinsulinemia, hyperglycemia, high levels of IGF-1 and inflammatory cytokines in common. Therefore,
these can be considered triggers for cancer development and growth. In addition, low-grade inflammation that
modulates the activation of immune cells, cellular metabolism, and production of cytokines and chemokines are
common in obesity, cancer, and insulin resistance. Pharmacological strategies are necessary when a change in
lifestyle does not improve glycemic homeostasis. In this regard, thiazolidinediones (TZD) possess multiple molecular
targets and regulate PPARγ in obesity and cancer related to insulin resistance, while metformin acts
through the AMPK pathway.
Objective:
The aim of this study was to review TZD and metformin as pharmacological treatments for insulin
resistance associated with obesity and cancer.
Conclusions:
Thiazolidinediones restored adiponectin secretion and leptin sensitivity, reduced lipid droplets in
hepatocytes and orexigen peptides in the hypothalamus. In cancer cells, TZD reduced proliferation, production of
reactive oxygen species, and inflammation by acting through the mTOR and NFκB pathways. Metformin has
similar effects, though these are AMPK-dependent. In addition, both drugs can be efficient against certain side
effects caused by chemotherapy.
Collapse
Affiliation(s)
- Luana A. Biondo
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Alexandre A. S. Teixeira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Karen C. de O. S. Ferreira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jose C. R. Neto
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Induction of peroxisome proliferator activated receptor γ (PPARγ) mediated gene expression and inhibition of induced nitric oxide production by Maerua subcordata (Gilg) DeWolf. BMC Complement Med Ther 2020; 20:80. [PMID: 32164648 PMCID: PMC7076844 DOI: 10.1186/s12906-020-2856-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The health benefits of botanicals is linked to their phytochemicals that often exert pleiotropic effects via targeting multiple molecular signaling pathways such as the peroxisome proliferator-activated receptors (PPARs) and the nuclear factor kappaB (NFκB). The PPARs are transcription factors that control metabolic homeostasis and inflammation while the NF-κB is a master regulator of inflammatory genes such as the inducible nitric-oxide synthase that result in nitric oxide (NO) overproduction. METHODS Extracts of Maerua subcordata (MS) and selected candidate constituents thereof, identified by liquid chromatography coupled to mass spectroscopy, were tested for their ability to induce PPARγ mediated gene expression in U2OS-PPARγ cells using luciferase reporter gene assay and also for their ability to inhibit lipopolysaccharide (LPS) induced NO production in RAW264.7 macrophages. While measuring the effect of test samples on PPARγ mediated gene expression, a counter assay that used U2OS-Cytotox cells was performed to monitor cytotoxicity or any non-specific changes in luciferase activity. RESULTS The results revealed that the fruit, root, and seed extracts were non-cytotoxic up to a concentration of 30 g dry weight per litre (gDW/L) and induced PPARγ mediated gene expression but the leaf extract showed some cytotoxicity and exhibited minimal induction. Instead, all extracts showed concentration (1-15 gDW/L) dependent inhibition of LPS induced NO production. The root extract showed weaker inhibition. Among the candidate constituents, agmatine, stachydrine, trigonelline, indole-3-carboxyaldehyde, plus ethyl-, isobutyl-, isopropyl, and methyl-isothiocyanates showed similar inhibition, and most showed increased inhibition with increasing concentration (1-100 μM) although to a lesser potency than the positive control, aminoguanidine. CONCLUSION The present study demonstrated for the first time the induction of PPARγ mediated gene expression by MS fruit, root, and seed extracts and the inhibition of LPS induced NO production by MS fruit, leaf, root, and seed extracts and some candidate constituents thereof.
Collapse
|
8
|
Wierenga KA, Wee J, Gilley KN, Rajasinghe LD, Bates MA, Gavrilin MA, Holian A, Pestka JJ. Docosahexaenoic Acid Suppresses Silica-Induced Inflammasome Activation and IL-1 Cytokine Release by Interfering With Priming Signal. Front Immunol 2019; 10:2130. [PMID: 31616405 PMCID: PMC6763728 DOI: 10.3389/fimmu.2019.02130] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Occupational exposure to respirable crystalline silica (cSiO2) has been etiologically linked to human autoimmunity. Intranasal instillation with cSiO2 triggers profuse inflammation in the lung and onset of autoimmunity in lupus-prone mice; however, dietary supplementation with the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) abrogates these responses. Inflammasome activation, IL-1 cytokine release, and death in alveolar macrophages following cSiO2 exposure are early and critical events that likely contribute to triggering premature autoimmune pathogenesis by this particle. Here we tested the hypothesis that DHA suppresses cSiO2-induced NLRP3 inflammasome activation, IL-1 cytokine release, and cell death in the macrophage. The model used was the murine macrophage RAW 264.7 cell line stably transfected with the inflammasome adapter protein ASC (RAW-ASC). Following priming with LPS, both the canonical activator nigericin and cSiO2 elicited robust inflammasome activation in RAW-ASC cells, as reflected by IL-1β release and caspase-1 activation. These responses were greatly diminished or absent in wild-type RAW cells. In contrast to IL-1β, cSiO2 induced IL-1α release in both RAW-ASC and to a lesser extent in RAW-WT cells after LPS priming. cSiO2-driven effects in RAW-ASC cells were confirmed in bone-marrow derived macrophages. Pre-incubating RAW-ASC cells with 10 and 25 μM DHA for 24 h enriched this fatty acid in the phospholipids by 15- and 25-fold, respectively, at the expense of oleic acid. DHA pre-incubation suppressed inflammasome activation and release of IL-1β and IL-1α by nigericin, cSiO2, and two other crystals - monosodium urate and alum. DHA's suppressive effects were linked to inhibition of LPS-induced Nlrp3, Il1b, and Il1a transcription, potentially through the activation of PPARγ. Finally, nigericin-induced death was inflammasome-dependent, indicative of pyroptosis, and could be inhibited by DHA pretreatment. In contrast, cSiO2-induced death was inflammasome-independent and not inhibited by DHA. Taken together, these findings indicate that DHA suppresses cSiO2-induced inflammasome activation and IL-1 cytokine release in macrophages by acting at the level of priming, but was not protective against cSiO2-induced cell death.
Collapse
Affiliation(s)
- Kathryn A Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Josephine Wee
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kristen N Gilley
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Lichchavi D Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Melissa A Bates
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Mikhail A Gavrilin
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH, United States
| | - Andrij Holian
- Department of Biomedical and Pharmaceutical Sciences, Center for Environmental Health Sciences, University of Montana, Missoula, MT, United States
| | - James J Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
9
|
He J, Qi D, Tang XM, Deng W, Deng XY, Zhao Y, Wang DX. Rosiglitazone promotes ENaC-mediated alveolar fluid clearance in acute lung injury through the PPARγ/SGK1 signaling pathway. Cell Mol Biol Lett 2019; 24:35. [PMID: 31160894 PMCID: PMC6540532 DOI: 10.1186/s11658-019-0154-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Background Pulmonary edema is one of the pathological characteristics of acute respiratory distress syndrome (ARDS). The epithelial sodium channel (ENaC) is thought to be the rate-limiting factor for alveolar fluid clearance (AFC) during pulmonary edema. The peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone was shown to stimulate ENaC-mediated salt absorption in the kidney. However, its role in the lung remains unclear. Here, we investigated the role of the PPARγ agonist in the lung to find out whether it can regulate AFC during acute lung injury (ALI). We also attempted to elucidate the mechanism for this. Methods Our ALI model was established through intratracheal instillation of lipopolysaccharide (LPS) in C57BL/6 J mice. The mice were randomly divided into 4 groups of 10. The control group underwent a sham operation and received an equal quantity of saline. The three experimental groups underwent intratracheal instillation of 5 mg/kg LPS, followed by intraperitoneal injection of 4 mg/kg rosiglitazone, 4 mg/kg rosiglitazone plus 1 mg/kg GW9662, or only equal quantity of saline. The histological morphology of the lung, the levels of TNF-α and IL-1β in the bronchoalveolar lavage fluid (BALF), the level of AFC, and the expressions of αENaC and serum and glucocorticoid-induced kinase-1 (SGK1) were determined. Type 2 alveolar (AT II) cells were incubated with rosiglitazone (15 μM) with or without GW9662 (10 μM). The expressions of αENaC and SGK1 were determined 24 h later. Results A mouse model of ALI was successfully established. Rosiglitazone significantly ameliorated the lung injury, decreasing the TNF-α and IL-1β levels in the BALF, enhancing AFC, and promoting the expressions of αENaC and SGK1 in ALI mice, which were abolished by the specific PPARγ blocker GW9662. In vitro, rosiglitazone increased the expressions of αENaC and SGK1. This increase was prevented by GW9662. Conclusions Rosiglitazone ameliorated the lung injury and promoted ENaC-mediated AFC via a PPARγ/SGK1-dependent signaling pathway, alleviating pulmonary edema in a mouse model of ALI.
Collapse
Affiliation(s)
- Jing He
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Di Qi
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Xu-Mao Tang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Wang Deng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Xin-Yu Deng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Yan Zhao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| | - Dao-Xin Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010 China
| |
Collapse
|
10
|
Edling CE, Fazmin IT, Chadda KR, Ahmad S, Valli H, Huang CLH, Jeevaratnam K. Atrial Transcriptional Profiles of Molecular Targets Mediating Electrophysiological Function in Aging and Pgc-1β Deficient Murine Hearts. Front Physiol 2019; 10:497. [PMID: 31068841 PMCID: PMC6491872 DOI: 10.3389/fphys.2019.00497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/08/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Deficiencies in the transcriptional co-activator, peroxisome proliferative activated receptor, gamma, coactivator-1β are implicated in deficient mitochondrial function. The latter accompanies clinical conditions including aging, physical inactivity, obesity, and diabetes. Recent electrophysiological studies reported that Pgc-1β-/- mice recapitulate clinical age-dependent atrial pro-arrhythmic phenotypes. They implicated impaired chronotropic responses to adrenergic challenge, compromised action potential (AP) generation and conduction despite normal AP recovery timecourses and background resting potentials, altered intracellular Ca2+ homeostasis, and fibrotic change in the observed arrhythmogenicity. OBJECTIVE We explored the extent to which these age-dependent physiological changes correlated with alterations in gene transcription in murine Pgc-1β-/- atria. METHODS AND RESULTS RNA isolated from murine atrial tissue samples from young (12-16 weeks) and aged (>52 weeks of age), wild type (WT) and Pgc-1β-/- mice were studied by pre-probed quantitative PCR array cards. We examined genes encoding sixty ion channels and other strategic atrial electrophysiological proteins. Pgc-1β-/- genotype independently reduced gene transcription underlying Na+-K+-ATPase, sarcoplasmic reticular Ca2+-ATPase, background K+ channel and cholinergic receptor function. Age independently decreased Na+-K+-ATPase and fibrotic markers. Both factors interacted to alter Hcn4 channel activity underlying atrial automaticity. However, neither factor, whether independently or interactively, affected transcription of cardiac Na+, voltage-dependent K+ channels, surface or intracellular Ca2+ channels. Nor were gap junction channels, β-adrenergic receptors or transforming growth factor-β affected. CONCLUSION These findings limit the possible roles of gene transcriptional changes in previously reported age-dependent pro-arrhythmic electrophysiologial changes observed in Pgc-1β-/- atria to an altered Ca2+-ATPase (Atp2a2) expression. This directly parallels previously reported arrhythmic mechanism associated with p21-activated kinase type 1 deficiency. This could add to contributions from the direct physiological outcomes of mitochondrial dysfunction, whether through reactive oxygen species (ROS) production or altered Ca2+ homeostasis.
Collapse
Affiliation(s)
- Charlotte E. Edling
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Ibrahim T. Fazmin
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom,Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Karan R. Chadda
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom,Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Haseeb Valli
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom,Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom,Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom,School of Medicine, Perdana University-Royal College of Surgeons in Ireland, Selangor, Malaysia,*Correspondence: Kamalan Jeevaratnam,
| |
Collapse
|
11
|
Hamblin MR. Novel pharmacotherapy for burn wounds: what are the advancements. Expert Opin Pharmacother 2019; 20:305-321. [PMID: 30517046 PMCID: PMC6364296 DOI: 10.1080/14656566.2018.1551880] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The prognosis for severe burns has improved significantly over the past 50 years. Meanwhile, burns have become an affliction mainly affecting the less well-developed regions of the world. Early excision and skin grafting has led to major improvements in therapeutic outcomes. AREAS COVERED The purpose of this article is to survey the use of pharmacotherapy to treat different pathophysiological complications of burn injury. The author, herein, discusses the use of drug treatments for a number of systemic metabolic disturbances including hyperglycemia, elevated catabolism, and gluconeogenesis. EXPERT OPINION Advancements in personalized and molecular medicine will make an impact on burn therapy. Similarities between severe burns and other critically ill patients will lead to cross-fertilization between different medical specialties. Furthermore, advances in stem cells and tissue regeneration will lead to improved healing and less lifelong disability. Indeed, research in new drug therapy for burns is actively progressing for many different complications.
Collapse
Affiliation(s)
- Michael R Hamblin
- a Wellman Center for Photomedicine , Massachusetts General Hospital , Boston , MA , USA
- b Department of Dermatology , Harvard Medical School , Boston , MA , USA
- c Harvard-MIT Division of Health Sciences and Technology , Cambridge , MA , USA
| |
Collapse
|
12
|
Li J, Shen X. Effect of rosiglitazone on inflammatory cytokines and oxidative stress after intensive insulin therapy in patients with newly diagnosed type 2 diabetes. Diabetol Metab Syndr 2019; 11:35. [PMID: 31073335 PMCID: PMC6499940 DOI: 10.1186/s13098-019-0432-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To evaluate the effect of insulin sensitizer on inflammatory cytokines and oxidative stress in patients with newly diagnosed type 2 diabetes mellitus (T2DM). METHODS After intensive insulin therapy, patients with newly diagnosed T2DM were continuously treated with either insulin sensitizer or insulin for 48 weeks, and then their inflammatory cytokine and oxidative stress levels were measured. RESULTS Tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, hypersensitive C reactive protein (hs-CRP), malondialdehyde (MDA), and 8-iso-prostaglandin F2α (8-iso-PGF2α) levels of the rosiglitazone (RSG) group and the rosiglitazone combined with metformin (RSG + metformin) group were significantly reduced after the treatments (P < 0.05). Hs-CRP, MDA, and 8-iso-PGF2α levels of the metformin group were significantly reduced after the treatments (P < 0.05). Superoxide dismutase (SOD) and total antioxidant capacity (TAC) were significantly increased after the treatments in all three groups (P < 0.05 and P < 0.01). CONCLUSION Early application of insulin sensitizers improved inflammation and oxidative stress in patients with newly diagnosed T2DM.
Collapse
Affiliation(s)
- Juan Li
- Department of Emergency, Zhongshan Hospital Xiamen University, Xiamen, 361004 Fujian China
| | - Xingping Shen
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, 361004 Fujian China
| |
Collapse
|
13
|
Chadda KR, Edling CE, Valli H, Ahmad S, Huang CLH, Jeevaratnam K. Gene and Protein Expression Profile of Selected Molecular Targets Mediating Electrophysiological Function in Pgc-1α Deficient Murine Atria. Int J Mol Sci 2018; 19:ijms19113450. [PMID: 30400228 PMCID: PMC6274828 DOI: 10.3390/ijms19113450] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 12/27/2022] Open
Abstract
Increases in the prevalence of obesity, insulin resistance, and metabolic syndrome has led to the increase of atrial fibrillation (AF) cases in the developed world. These AF risk factors are associated with mitochondrial dysfunction, previously modelled using peroxisome proliferator activated receptor-γ (PPARγ) coactivator-1 (Pgc-1)-deficient murine cardiac models. We explored gene and protein expression profiles of selected molecular targets related to electrophysiological function in murine Pgc-1α−/− atria. qPCR analysis surveyed genes related to Na+-K+-ATPase, K+ conductance, hyperpolarisation-activated cyclic nucleotide-gated (Hcn), Na+ channels, Ca2+ channels, and indicators for adrenergic and cholinergic receptor modulation. Western blot analysis for molecular targets specific to conduction velocity (Nav1.5 channel and gap junctions) was performed. Transcription profiles revealed downregulation of molecules related to Na+-K+-ATPase transport, Hcn-dependent pacemaker function, Na+ channel-dependent action potential activation and propagation, Ca2+ current generation, calsequestrin-2 dependent Ca2+ homeostasis, and adrenergic α1D dependent protection from hypertrophic change. Nav1.5 channel protein expression but not gap junction expression was reduced in Pgc-1α−/− atria compared to WT. Nav1.5 reduction reflects corresponding reduction in its gene expression profile. These changes, as well as the underlying Pgc-1α−/− alteration, suggest potential pharmacological targets directed towards either upstream PGC-1 signalling mechanisms or downstream ion channel changes.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Charlotte E Edling
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
| | - Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge CB2 1QW, UK.
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
- School of Medicine, Perdana University-Royal College of Surgeons Ireland, Serdang 43400, Malaysia.
| |
Collapse
|
14
|
Cho R, Yang C, Tseng H, Hsiao L, Lin C, Yang C. Haem oxygenase-1 up-regulation by rosiglitazone via ROS-dependent Nrf2-antioxidant response elements axis or PPARγ attenuates LPS-mediated lung inflammation. Br J Pharmacol 2018; 175:3928-3946. [PMID: 30088830 PMCID: PMC6151343 DOI: 10.1111/bph.14465] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Haem oxygenase-1 (HO-1) is induced by thiazolidinediones including rosiglitazone and exerts anti-inflammatory effects in various models. However, the molecular mechanisms underlying rosiglitazone-induced HO-1 expression remain largely unknown in human pulmonary alveolar epithelial cells (HPAEpiCs). EXPERIMENTAL APPROACH HO-1 expression was determined by real time-PCR, Western blotting and promoter reporter analyses. Signalling pathways were investigated using pharmacological inhibitors or specific siRNAs. Interactions between nuclear factor erythroid-2-related factor (Nrf2) and antioxidant response elements (ARE) binding site of the HO-1 promoter were investigated with chromatin immunoprecipitation assays. KEY RESULTS Up-regulation of HO-1 in HPAEpiCs or in mice by rosiglitazone blunted ICAM-1 expression and monocyte adhesion to HPAEpiCs challenged with LPS. Rosiglitazone-induced HO-1 expression was significantly attenuated by NADPH oxidase (NOX) inhibitors (apocynin and diphenyleneiodonium) or ROS scavenger (N-acetyl cysteine). The involvement of NOX activity and ROS generation in rosiglitazone-induced HO-1 expression was confirmed by transfection with p47phox or NOX2 siRNA. Moreover, pretreatment with the inhibitors of c-Src (c-Srci II), proline-rich tyrosine kinase 2 (Pyk2) (PF431396), Akt (Akti VIII) or PPARγ (GW9662) and transfection with siRNA of c-Src, Pyk2, Akt or PPARγ abolished the rosiglitazone-induced HO-1 expression in HPAEpiCs. Subsequently, Nrf2 was activated by phosphorylation of c-Src, Pyk2 and Akt, which turned on transcription of HO-1 gene by binding to AREs binding site and enhancing ARE promoter activity. CONCLUSIONS AND IMPLICATIONS Rosiglitazone induces HO-1 expression via either NOX/ROS/c-Src/Pyk2/Akt-dependent Nrf2 activation or PPARγ in HPAEpiCs and suppresses LPS-mediated inflammatory responses, suggesting that PPARγ agonists may be useful for protection against pulmonary inflammation.
Collapse
Affiliation(s)
- Rou‐Ling Cho
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of MedicineChang Gung UniversityTao‐YuanTaiwan
| | - Chien‐Chung Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of MedicineChang Gung UniversityTao‐YuanTaiwan
- Department of Traditional Chinese MedicineChang Gung Memorial Hospital at Tao‐YuanTao‐YuanTaiwan
| | - Hui‐Ching Tseng
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of MedicineChang Gung UniversityTao‐YuanTaiwan
| | - Li‐Der Hsiao
- Department of AnestheticsChang Gung Memorial Hospital at Linkuo and Chang Gung UniversityTao‐YuanTaiwan
| | - Chih‐Chung Lin
- Department of AnestheticsChang Gung Memorial Hospital at Linkuo and Chang Gung UniversityTao‐YuanTaiwan
| | - Chuen‐Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of MedicineChang Gung UniversityTao‐YuanTaiwan
- Department of AnestheticsChang Gung Memorial Hospital at Linkuo and Chang Gung UniversityTao‐YuanTaiwan
- Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human EcologyChang Gung University of Science and TechnologyTao‐YuanTaiwan
| |
Collapse
|
15
|
Chistyakov DV, Azbukina NV, Lopachev AV, Kulichenkova KN, Astakhova AA, Sergeeva MG. Rosiglitazone as a Modulator of TLR4 and TLR3 Signaling Pathways in Rat Primary Neurons and Astrocytes. Int J Mol Sci 2018; 19:E113. [PMID: 29301276 PMCID: PMC5796062 DOI: 10.3390/ijms19010113] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 01/06/2023] Open
Abstract
An antidiabetic drug of the thiazolidinedione class, rosiglitazone (RG) demonstrates anti-inflammatory properties in various brain pathologies. The mechanism of RG action in brain cells is not fully known. To unravel mechanisms of RG modulation of toll-like receptor (TLR) signaling pathways, we compare primary rat neuron and astrocyte cultures stimulated with the TLR4 agonist lipopolysaccharide (LPS) and the TLR3 agonist poly I:C (PIC). Both TLR agonists induced tumor necrosis factor (TNFα) release in astrocytes, but not in neurons. Neurons and astrocytes released interleukin-10 (IL-10) and prostaglandin E2 (PGE₂) in response to LPS and PIC. RG decreased TLR-stimulated TNFα release in astrocytes as well as potentiated IL-10 and PGE₂ release in both astrocytes and neurons. RG induced phosphorylation of p38 and JNK MAPK (mitogen-activated protein kinase) in neurons. The results reveal new role of RG as a modulator of resolution of neuroinflammation.
Collapse
Affiliation(s)
- Dmitry V Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
- Laboratory of electrophysiology, Pirogov Russian National Research Medical University, Moscow 117997, Russia.
| | - Nadezda V Azbukina
- Faculty of Bioengineering and Bioinformatics, Moscow Lomonosov State University, Moscow 119234 Russia.
| | | | | | - Alina A Astakhova
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
| | - Marina G Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119992, Russia.
| |
Collapse
|
16
|
Flori E, Rosati E, Cardinali G, Kovacs D, Bellei B, Picardo M, Maresca V. The α-melanocyte stimulating hormone/peroxisome proliferator activated receptor-γ pathway down-regulates proliferation in melanoma cell lines. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:142. [PMID: 29020973 PMCID: PMC5637056 DOI: 10.1186/s13046-017-0611-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022]
Abstract
Background The α-Melanocyte Stimulating Hormone (αMSH)/Melanocortin-1 receptor (MC1R) interaction promotes melanogenesis through the cAMP/PKA pathway. The direct induction of this pathway by Forskolin (FSK) is also known to enhance melanocyte proliferation. αMSH acts as a mitogenic agent in melanocytes and its effect on proliferation of melanoma cells is less known. We previously identified the αMSH/Peroxisome Proliferator Activated Receptor (PPARγ) pathway as a new pathway on the B16-F10 mouse melanoma cell line. αMSH induced the translocation of PPARγ into the nucleus as an active transcription factor. This effect was independent of the cAMP/PKA pathway and was mediated by the activation of the PI(4,5)P2/PLC pathway, a pathway which we have described to be triggered by the αMSH-dependent MC1R stimulation. Moreover, in the same study, preliminary experiments showed that mouse melanoma cells responded to αMSH by reducing proliferation and that PPARγ was involved in this effect. Due to its key role in the control of cell proliferation, PPARγ agonists are used in therapeutic models for different forms of cancer, including melanoma. The purpose of this study was: (a) to confirm the different proliferative behavior in response to αMSH in healthy and in melanoma condition; (b) to verify whether the cAMP/PKA pathway and the PLC/PPARγ pathway could exert an antagonistic function in the control of proliferation; (c) to deepen the knowledge of the molecular basis responsible for the down-proliferative response of melanoma cells after exposure to αMSH. Methods We employed B16-F10 cell line, a human melanoma cell line (Mel 13) and two primary cultures of human melanocytes (NHM 1 and NHM 2, respectively), all expressing a wild type MC1R and responding to the αMSH in terms of pigmentation. We evaluated cell proliferation through: a) cell counting, b) cell cycle analysis c) protein expression of proliferation modulators (p27, p21, cyclin D1 and cyclin E). Results The αMSH acted as a mitogenic agent in primary cultures of human melanocytes, whereas it determined a slow down of proliferation in melanoma cell lines. FSK, as an inducer of the cAMP/PKA pathway, reproduced the αMSH mediated effect on proliferation in NHMs but it did not mimic the αMSH effect on proliferation in B16-F10 and Mel 13 melanoma cell lines. Meanwhile, 3 M3-FBS (3 M3), as an inducer of PI(4,5)P2/PLC pathway, reproduced the αMSH proliferative effect. Further experiments, treating melanoma cell lines with αMSH in the presence/absence of GW9662, as an inhibitor of PPARγ, confirmed the key role of this transcription factor in decreasing cell proliferation in response to the hormone exposure. Conclusions In both melanoma cell lines, αMSH determined the reduction of proliferation through the PI(4,5)P2/PLC pathway, employing PPARγ as an effector element. These evidence could offer perspectives for new therapeutic approaches for melanoma. Electronic supplementary material The online version of this article (10.1186/s13046-017-0611-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Enrica Flori
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Eleonora Rosati
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Giorgia Cardinali
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Daniela Kovacs
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy
| | - Vittoria Maresca
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute (IRCCS), Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
17
|
The role of pparγ and autophagy in ros production, lipid droplets biogenesis and its involvement with colorectal cancer cells modulation. Cancer Cell Int 2017; 17:82. [PMID: 28932171 PMCID: PMC5603033 DOI: 10.1186/s12935-017-0451-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022] Open
Abstract
Background In cancer cells, autophagy can act as both tumor suppressor, when autophagic event eliminates cellular contends which exceeds the cellular capacity of regenerate promoting cell death, and as a pro-survival agent removing defective organelles and proteins and helping well-established tumors to maintain an accelerated metabolic state while still dealing with harsh conditions, such as inflammation. Many pathways can coordinate the autophagic process and one of them involves the transcription factors called PPARs, which also regulate cellular differentiation, proliferation and survival. The PPARγ activation and autophagy initiation seems to be interrelated in a variety of cell types. Methods Caco-2 cells were submitted to treatment with autophagy and PPARγ modulators and the relationship between both pathways was determined by western blotting and confocal microscopy. The effects of such modulations on Caco-2 cells, such as lipid bodies biogenesis, cell death, proliferation, cell cycle, ROS production and cancer stem cells profiling were analyzed by flow cytometry. Results PPARγ and autophagy pathways seem to be overlap in Caco-2 cells, modulating each other in different ways and determining the lipid bodies biogenesis. In general, inhibition of autophagy by 3-MA leaded to reduced cell proliferation, cell cycle arrest and, ultimately, cell death by apoptosis. In agreement with these results, ROS production was increased in 3-MA treated cells. Autophagy also seems to play an important role in cancer stem cells profiling. Rapamycin and 3-MA induced epithelial and mesenchymal phenotypes, respectively. Conclusions This study helps to elucidate in which way the induction or inhibition of these pathways regulate each other and affect cellular properties, such as ROS production, lipid bodies biogenesis and cell survive. We also consolidate autophagy as a key factor for colorectal cancer cells survival in vitro, pointing out a potential side effect of autophagic inhibition as a therapeutic application for this disease and demonstrate a novel regulation of PPARγ expression by inhibition of PI3K III. Electronic supplementary material The online version of this article (doi:10.1186/s12935-017-0451-5) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Lee JW, Mok HJ, Lee DY, Park SC, Kim GS, Lee SE, Lee YS, Kim KP, Kim HD. UPLC-QqQ/MS-Based Lipidomics Approach To Characterize Lipid Alterations in Inflammatory Macrophages. J Proteome Res 2017; 16:1460-1469. [DOI: 10.1021/acs.jproteome.6b00848] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jae Won Lee
- Department
of Applied Chemistry, The Institute of Natural Science, College of
Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Hyuck Jun Mok
- Department
of Applied Chemistry, The Institute of Natural Science, College of
Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Dae Young Lee
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Seung Cheol Park
- Department
of Applied Chemistry, The Institute of Natural Science, College of
Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Geum-Soog Kim
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Seung-Eun Lee
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Young-Seob Lee
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Kwang Pyo Kim
- Department
of Applied Chemistry, The Institute of Natural Science, College of
Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyung Don Kim
- Department
of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
- Department
of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
19
|
Xiang S, Zeng Y, Xiong B, Qin Y, Huang X, Jiang Y, Luo W, Sooranna SR, Pinhu L. Transforming growth factor beta 1 induced endothelin-1 release is peroxisome proliferator-activated receptor gamma dependent in A549 cells. JOURNAL OF INFLAMMATION-LONDON 2016; 13:19. [PMID: 27293383 PMCID: PMC4902962 DOI: 10.1186/s12950-016-0128-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/07/2016] [Indexed: 01/30/2023]
Abstract
Background Endothelin-1 (ET-1) is involved in pulmonary vascular remodeling. The aim of this study was to investigate the biochemical interactions between PPAR-γ, TGF-β1 and ET-1 in vitro. Methods A549 cells were pre-treated with S2505 (10 μM), S2871 (10 μM) with/without SB203580 (10 μM) for 60 min following 2 h treatment with 10 ng/mL TGF-β1. A549 cells were also transfected with positive or negative PPAR-γ plasmids for comparison. RT-PCR, ELISA, western blotting and confocal laser scanning microscopy (CLSM) were used to measure the relevant expression of mRNA, protein, mediators of pathways and nuclear factor translocation. Results SB203580 inhibited TGF-β1 induced ET-1 expression in A549 cells. S2871 decreased PPAR-γ mRNA and increase TGF-β1-induced ET-1 expression. S2871 increased phosphorylation of p38 MAPK and Smad2. Cells transfected with PPAR-γ negative plasmid increased TGF-β1 induced ET-1 expression, and increased the expression of phospho-p38 MAPK and phospho-Smad2. S2505 increased PPAR-γ mRNA expression, suppressed the increased TGF-β1-induced expression of ET-1. S2505 inhibited TGF-β1 induced phosphorylation of p38 MAPK and Smad2, also the nuclear translocation of Smad2. Cells transfected with PPAR-γ positive plasmid reduced TGF-β1-induced ET-1 expression, and inhibited the expression of phospho-p38 MAPK and phospho-Smad2. Conclusions TGF-β1 induced release of endothelin-1 is PPAR-γ dependent in cultured A549 cells.
Collapse
Affiliation(s)
- Shulin Xiang
- The First Clinical Medical College of Jinan University, Guangzhou, 510630 Guangdong Province China.,Department of Intensive Care Unit, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021 China
| | - Yi Zeng
- Department of Central Laboratory, Youjiang Medical University for Nationalities, Baise, 533000 Guangxi Zhuang Autonomous Region China
| | - Bin Xiong
- Department of Intensive Care Unit, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021 China
| | - Yueqiu Qin
- Department of Digestive Medicine, Youjiang Medical University for Nationalities, Baise, 533000 Guangxi Zhuang Autonomous Region China
| | - Xia Huang
- The First Clinical Medical College of Jinan University, Guangzhou, 510630 Guangdong Province China.,Department of Respiratory Medicine, Youjiang Medical University for Nationalities, Baise, 533000 Guangxi Zhuang Autonomous Region China
| | - Yujie Jiang
- The First Clinical Medical College of Jinan University, Guangzhou, 510630 Guangdong Province China.,Department of Respiratory Medicine, Youjiang Medical University for Nationalities, Baise, 533000 Guangxi Zhuang Autonomous Region China
| | - Weigui Luo
- Department of Respiratory Medicine, 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, Youjiang Medical University for Nationalities, Baise, 533000 Guangxi Zhuang Autonomous Region China
| |
Collapse
|
20
|
Zhang X, Zhang R, Lv P, Yang J, Deng Y, Xu J, Zhu R, Zhang D, Yang Y. Emodin up-regulates glucose metabolism, decreases lipolysis, and attenuates inflammation in vitro. J Diabetes 2015; 7:360-8. [PMID: 24981886 DOI: 10.1111/1753-0407.12190] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/15/2014] [Accepted: 06/22/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Emodin, the major bioactive component of Rheum palmatum, has many different activities, including antitumor, anti-inflammatory, and antidiabetes effects. Recently, emodin was reported to regulate energy metabolism. In the present study, we further explored the effects of emodin on glucose and lipid metabolism. METHODS Differentiated C2C12 myotubes and 3T3-L1 adipocytes were treated with or without different concentrations of emodin (6.25, 12.5, 25 or 50 μmol/L) for different time (1 h, 3 h, 12 h, 24 h or 48 h). Glucose metabolism, oxygen consumption, lactic acid levels, glycerol levels, and inflammation pathways were then evaluated. Cells were collected for quantitative polymerase chain reaction (PCR) and western blot analysis. RESULTS Emodin upregulated glucose uptake and consumption in both C2C12 myotubes and 3T3-L1 adipocytes, with glycolysis increased. Furthermore, emodin inhibited lipolysis under basal conditions (as well as in the presence of 10 ng/ml tumor necrosis factor (TNF-)-α in 3T3-L1 adipocytes) and significantly decreased phosphorylated perilipin. Moreover, emodin inhibited the nuclear factor-κB and extracellular signal-regulated kinase pathways in C2C12 myotubes and 3T3-L1 adipocytes. CONCLUSIONS Emodin upregulates glucose metabolism, decreases lipolysis, and inhibits inflammation in C2C12 myotubes and 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|