1
|
Chen J, Chen Y, Huang R, Zhang P, Huo Z, Li Y, Xiao H, Guan H, Li H. Long noncoding RNA FAM111A-DT promotes aggressiveness of papillary thyroid cancer via activating NF-κB signaling. J Endocrinol Invest 2025; 48:1121-1136. [PMID: 39786708 DOI: 10.1007/s40618-025-02531-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Long noncoding RNAs (lncRNAs) play crucial regulatory roles in the tumorigenesis and progression of various cancers. However, the functional roles of lncRNAs in papillary thyroid cancer (PTC) remain unclear. In this study, we investigated the functional role of the lncRNA FAM111A-DT in PTC progression and the underlying mechanisms. METHODS Different expression levels of lncRNAs in PTC were compared via analysis of the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. Bioinformatics analyses and qRT‒PCR were used to investigate the expression of FAM111A-DT in PTC. Cell proliferation was measured via CCK8, EdU, colony formation, and flow cytometry assays. Cell migration and invasion were examined by wound healing and Transwell assays. Apoptosis was detected via flow cytometry. RNA sequencing, qRT‒PCR, Western blot, immunofluorescence and dual-luciferase reporter assays were performed to assess the underlying mechanisms involved. RESULTS FAM111A-DT was highly expressed in PTC and associated with poor prognosis, thyroid dedifferentiation, various clinical features and the BRAFV600E mutation in PTC patients. Overexpression of FAM111A-DT enhanced the proliferation, migration and invasion of PTC cells while reducing their degree of apoptosis. The NF-κB signaling pathway was activated in FAM111A-DT-overexpressing PTC cells. The NF-κB inhibitor PDTC attenuated the promotive effects of FAM111A-DT on aggressive phenotypes and NF-κB pathway activity in PTC cells. CONCLUSION FAM111A-DT is upregulated in PTC, and its expression is associated with poor clinical outcomes. FAM111A-DT plays an oncogenic role by, at least partially, activating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Junxin Chen
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yue Chen
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rong Huang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pengyuan Zhang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zijun Huo
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haipeng Xiao
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongyu Guan
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Hai Li
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Department of Endocrinology, Guizhou Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guizhou, China.
| |
Collapse
|
2
|
Li Y, Li J, Liu X, Cheng Z, Gao N, Kang J, Wang X. Polygonatum sibiricum Polysaccharides Alleviate LPS-Induced Liver Injury in Chicks by Suppressing Inflammation and Oxidative Stress Through the PPAR Signaling Pathway. Antioxidants (Basel) 2025; 14:418. [PMID: 40298760 PMCID: PMC12024365 DOI: 10.3390/antiox14040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Polygonatum sibiricum polysaccharides (PSPs), plant-derived polysaccharides widely used in the pharmaceutical field, exhibit various biological activities, including anti-inflammatory and antioxidant effects. However, research on their application in chicks remains limited. Therefore, the aim of this study is to investigate the protective mechanism of PSP against liver injury in chicks using an LPS-induced inflammatory model. A total of 200 one-day-old Hy-Line Brown laying chicks were randomly assigned to five groups (40 chicks each): a control group (CON) fed a basal diet, an LPS group, and three PSP groups receiving low (250 mg/L), medium (500 mg/L), and high (1000 mg/L) doses of PSP (PSP250_LPS, PSP500_LPS, and PSP1000_LPS, respectively). The experiment lasted 21 days. During this period, the LPS and PSP groups were intraperitoneally injected with 1500 μg/kg LPS on days 14, 16, 18, and 20, while the CON group received normal saline. On day 21, organs were collected for analysis. The results indicated that PSP treatment significantly reduced the liver and kidney indices that were elevated by LPS (p < 0.05) without affecting the indices of the spleen, thymus, or bursa of Fabricius (p > 0.05). Histological analysis revealed that PSP alleviated LPS-induced ballooning degeneration and cell swelling in hepatocytes. Furthermore, PSP treatment decreased the levels of ALT and AST and significantly mitigated increases in the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α while enhancing the level of the anti-inflammatory cytokine IL-10 (p < 0.05). Transcriptome sequencing of liver samples revealed that LPS significantly altered the expression of 10 genes in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which were regulated by PSP intervention. qPCR validation supported these findings. Furthermore, biochemical analyses of liver tissue showed that PSP alleviated oxidative stress by affecting levels of SOD, MDA, NADPH, ROS, and H2O2. In conclusion, PSP may alleviate LPS-induced liver injury in chicks by modulating the PPAR signaling pathway. These findings provide valuable insights for promoting healthy chick rearing and ensuring the safe supply of poultry products.
Collapse
Affiliation(s)
- Yang Li
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Jian Li
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Xiaowang Liu
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Zhili Cheng
- Hebei Provincial Animal Husbandry Station, Shijiazhuang 050011, China; (J.L.); (X.L.); (Z.C.)
| | - Nana Gao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Jungang Kang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| | - Xiaodan Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (Y.L.); (N.G.); (J.K.)
| |
Collapse
|
3
|
Miyata R, Suzuki M, Okazaki Y, Abe D, Nakajima Y. Peroxisome Proliferator-Activated Receptor-Gamma Activation by an Active Compound in Lythrum anceps (Koehne) Makino. J Cell Biochem 2025; 126:e70009. [PMID: 39980354 DOI: 10.1002/jcb.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Lythrum anceps (Koehne) Makino (Japanese common name: "Misohagi") is an edible plant belonging to the Lythraceae family. It is mainly distributed in Asia, Northern Africa, and Europe. Plants of the genus Lythrum exhibit a broad range of biological activities including anti-inflammatory and antimicrobial activities. Because of this, the plants are used in traditional medicine to treat hemorrhage, infected wounds, and dysentery. The activation of peroxisome proliferator-activated receptor-gamma (PPARγ) is an effective target for improving insulin resistance and anti-inflammatory activity. However, PPARγ activation by the genus Lythrum remains unclear. Aiming to evaluate PPARγ activation by L. anceps, we generated a reporter cell line using an artificial chromosome vector that stably expresses dual-color beetle luciferases. Dual-color real-time bioluminescence monitoring revealed marked PPARγ activation in L. anceps extracts. Moreover, ellagic acid was identified as a PPARγ activator present in L. anceps by a bioassay-guided fractionation approach.
Collapse
Affiliation(s)
- Ryo Miyata
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| | - Masanobu Suzuki
- Resources and Environment Division, Kochi Prefectural Industrial Technology Center, Kochi, Kochi, Japan
| | - Yuka Okazaki
- Resources and Environment Division, Kochi Prefectural Industrial Technology Center, Kochi, Kochi, Japan
| | - Daigo Abe
- Western Region Agricultural Research Center, National Agricultural and Food Research Organization (NARO), Zentsuji, Kagawa, Japan
| | - Yoshihiro Nakajima
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu, Kagawa, Japan
| |
Collapse
|
4
|
Liu J, Liu X, Guo L, Liu X, Gao Q, Wang E, Dong Z. PPARγ agonist alleviates calcium oxalate nephrolithiasis by regulating mitochondrial dynamics in renal tubular epithelial cell. PLoS One 2024; 19:e0310947. [PMID: 39325731 PMCID: PMC11426502 DOI: 10.1371/journal.pone.0310947] [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: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Kidney stone formation is a common disease that causes a significant threat to human health. The crystallization mechanism of calcium oxalate, the most common type of kidney stone, has been extensively researched, yet the damaging effects and mechanisms of calcium oxalate crystals on renal tubular epithelial cells remain incompletely elucidated. Regulated mitochondrial dynamics is essential for eukaryotic cells, but its role in the occurrence and progression of calcium oxalate (CaOx) nephrolithiasis is not yet understood. METHODS An animal model of calcium oxalate-related nephrolithiasis was established in adult male Sprague‒Dawley (SD) rats by continuously administering drinking water containing 1% ethylene glycol for 28 days. The impact of calcium oxalate crystals on mitochondrial dynamics and apoptosis in renal tubular epithelial cells was investigated using HK2 cells in vitro. Blood samples and bilateral kidney tissues were collected for histopathological evaluation and processed for tissue injury, inflammation, fibrosis, oxidative stress detection, and mitochondrial dynamics parameter analysis. RESULTS Calcium oxalate crystals caused higher levels of mitochondrial fission and apoptosis in renal tubular epithelial cells both in vivo and in vitro. Administration of a PPARγ agonist significantly alleviated mitochondrial fission and apoptosis in renal tubular epithelial cells, and improved renal function, accompanied by reduced levels of oxidative stress, increased antioxidant enzyme expression, alleviation of inflammation, and reduced fibrosis in vivo. CONCLUSION Our results indicated that increased mitochondrial fission in renal tubular epithelial cells is a critical component of kidney injury caused by calcium oxalate stones, leading to the accumulation of reactive oxygen species within the tissue and the subsequent initiation of apoptosis. Regulating mitochondrial dynamics represents a promising approach for calcium oxalate nephrolithiasis.
Collapse
Affiliation(s)
- Junfa Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xingyang Liu
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Xiongfei Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Zhitao Dong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Lai YH, Wu TC, Tsai BY, Hung YP, Lin HJ, Tsai YS, Ko WC, Tsai PJ. Peroxisome proliferator-activated receptor-γ as the gatekeeper of tight junction in Clostridioides difficile infection. Front Microbiol 2022; 13:986457. [PMID: 36439832 PMCID: PMC9691888 DOI: 10.3389/fmicb.2022.986457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/21/2022] [Indexed: 08/27/2023] Open
Abstract
Clostridioides difficile is a major causative pathogen of nosocomial antibiotic-associated diarrhea and severe colitis. Despite the use of vancomycin and fidaxomicin as standard drugs for the treatment of C. difficile infection (CDI), clinical relapse rates remain high. Therefore, new alternative therapeutics to treat CDI are urgently required. The nuclear receptor, peroxisome proliferator-activated receptor-γ (PPAR-γ), is mainly expressed in the adipose tissue and modulates lipid metabolism and insulin sensitization. Previous studies have shown that PPAR-γ is highly expressed in colonic tissues and regulates tight junction function in epithelial cells. However, the role of PPAR-γ in CDI pathogenesis remains unclear. In this study, we used a mouse model of CDI and found that both expression levels of PPAR-γ and the tight junction protein, occludin, were decreased in colonic tissues. Furthermore, to investigate the role of PPAR-γ in CDI, we used PPAR-γ defective mice and found that intestinal permeability and bacterial dissemination in these mice were significantly higher than those in wild-type mice during CDI. Administration of the PPAR-γ agonist, pioglitazone, to activate PPAR-γ activity improved the phenotypes of CDI, including bodyweight loss, inflammation, and intestinal integrity. Taken together, these results demonstrate that PPAR-γ is a potential therapeutic target in CDI, as it modulates colonic inflammation and integrity.
Collapse
Affiliation(s)
- Yi-Hsin Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tai-Chieh Wu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bo-Yang Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Pin Hung
- Departments of Internal Medicine, Tainan Hospital, Ministry of Health & Welfare, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Ju Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
- Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
6
|
Feng J, Li S, Zhang B, Duan N, Zhou R, Yan S, Elango J, Liu N, Wu W. FGFC1 Exhibits Anti-Cancer Activity via Inhibiting NF-κB Signaling Pathway in EGFR-Mutant NSCLC Cells. Mar Drugs 2022; 20:md20010076. [PMID: 35049931 PMCID: PMC8781927 DOI: 10.3390/md20010076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
FGFC1, an active compound isolated from the culture of marine fungi Stachybotrys longispora FG216, elicits fibrinolytic, anti-oxidative, and anti-inflammatory activity. We have previously reported that FGFC1 inhibited the proliferation, migration, and invasion of the non-small cell lung cancer (NSCLC) cells in vitro. However, the precise mechanisms of FGFC1 on NSCLC and its anti-cancer activity in vivo remains unclear. Hence, this study was focused to investigate the effects and regulatory mechanisms of FGFC1 on two NSCLC cell lines, EGFR-mutant PC9 (ex19del) and EGFR wild-type H1299. Results suggested that FGFC1 significantly inhibited proliferation, colony formation, as well as triggered G0/G1 arrest and apoptosis of PC9 cells in a dose- and time-dependent manner, but no obvious inhibitory effects were observed in H1299 cells. Subsequently, transcriptome analysis revealed that FGFC1 significantly down-regulated 28 genes related to the NF-κB pathway, including IL-6, TNF-α, and ICAM-1 in the PC9 cells. We further confirmed that FGFC1 decreased the expression of protein p-IKKα/β, p-p65, p-IκB, IL-6, and TNF-α. Moreover, NF-κB inhibitor PDTC could strengthen the effects of FGFC1 on the expression of CDK4, Cyclin D1, cleaved-PARP-1, and cleaved-caspase-3 proteins, suggesting that the NF-κB pathway plays a major role in FGFC1-induced cell cycle arrest and apoptosis. Correspondingly, the nuclear translocation of p-p65 was also suppressed by FGFC1 in PC9 cells. Finally, the intraperitoneal injection of FGFC1 remarkably inhibited PC9 xenograft growth and decreased the expression of Ki-67, p-p65, IL-6, and TNF-α in tumors. Our results indicated that FGFC1 exerted anti-cancer activity in PC9 cells via inhibiting the NF-κB signaling pathway, providing a possibility for FGFC1 to be used as a lead compound for the treatment of NSCLC in the future.
Collapse
Affiliation(s)
- Jingwen Feng
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Songlin Li
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai 201306, China;
| | - Bing Zhang
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Namin Duan
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Rui Zhou
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Shike Yan
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Jeevithan Elango
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
| | - Ning Liu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
- Engineering Research Center of Aquatic Product Processing & Preservation, Shanghai Ocean University, Shanghai 201306, China
- Correspondence: (N.L.); (W.W.)
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (J.F.); (B.Z.); (N.D.); (R.Z.); (S.Y.); (J.E.)
- Engineering Research Center of Aquatic Product Processing & Preservation, Shanghai Ocean University, Shanghai 201306, China
- Correspondence: (N.L.); (W.W.)
| |
Collapse
|
7
|
Panax notoginseng saponins induce apoptosis in retinoblastoma Y79 cells via the PI3K/AKT signalling pathway. Exp Eye Res 2022; 216:108954. [DOI: 10.1016/j.exer.2022.108954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/14/2021] [Accepted: 01/17/2022] [Indexed: 12/18/2022]
|
8
|
Preliminary evaluation of anticancer efficacy of pioglitazone combined with celecoxib for the treatment of non-small cell lung cancer. Invest New Drugs 2021; 40:1-9. [PMID: 34341904 DOI: 10.1007/s10637-021-01158-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Among the lung cancer types, non-small cell lung cancer (NSCLC) is prominent and less responsive to chemotherapy. The current chemotherapeutics for NSCLC are associated with several dose-limiting side effects like bone-marrow suppression, neurotoxicity, nephrotoxicity, and ototoxicity, etc. which are causing non-compliance in patients. Many tumors, including breasts, lung, ovarian, etc. overexpress PPAR-γ receptors and COX-2 enzymes, which play a crucial role in tumor progression, angiogenesis, and metastasis. Lack of PPAR-γ activation and overproduction of prostaglandins, result in uncontrolled activation of Ras/Raf/Mek ultimately, NF-κB mediated tumor proliferation. This study aimed to investigate the anti-cancer potential of PPAR-γ agonist Pioglitazone combined with COX-2 inhibitor Celelcoxib in NSCLC. METHODS Sixty adult Balb/C male mice were classified into sham control, disease control, and treatment groups. Mice were treated with Nicotine-derived nitrosamine ketone (NNK) (10 mg/kg), pioglitazone (10 & 20 mg/kg) and celecoxib (25 & 50 mg/kg). Weekly body weight, food intake, mean survival time & % increased life span were determined. Tumor weight and histopathological analysis were performed at the end of the study. RESULTS The significant tumor reducing potential of pioglitazone combined with celecoxib was observed (p < 0.05). The treatment groups (treated with pioglitazone and celecoxib) showed a remarkable decrease in lung tumor weight, improved life span and mean survival time (p < 0.05). Histopathological studies confirm that treatment groups (treated with pioglitazone and celecoxib) reframed the lung architecture compared to disease control. CONCLUSION Preliminary results revealed that pioglitazone adjunacy with celecoxib may be an effective chemo-preventive agent against NNK induce NSCLC.
Collapse
|
9
|
Kumari GK, Kiran AVVVR, Krishnamurthy PT. Preliminary evaluation on the beneficial effects of pioglitazone in the treatment of endometrial cancer. Med Oncol 2021; 38:71. [PMID: 34008039 DOI: 10.1007/s12032-021-01521-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
Endometrial cancer (EMC) is one of the complicated gynecological cancers, affecting more than three million women worldwide. Anticancer strategies such as chemotherapy, radiation, and surgery are found to be ineffective and are associated with patient incompliances. The aim of the present study is to repurpose non-oncological drug, i.e., Pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, in the treatment of endometrial cancer. The study groups consist of 50 female Swiss albino mice, out of which 40 had endometrial cancer induced with N-ethyl-N-nitrosourea (ENU) and estradiol hexadrobenzoate (EHB). The other groups received saline, EHB, paclitaxel, and different test doses of pioglitazones. Different preliminary parameters such as weekly body weight, mean survival time, percentage increase in life span, and uterine tissue weight were analyzed along with histopathological analysis. We observed a significant change in weekly body weight, improvement in percentage life span, and partial restoration of uterine tissue weight to normal compared to a standard drug, paclitaxel. In the present preliminary evaluation, we have identified that pioglitazone exhibited a significant dose-dependent anticancer activity against ENU- and EHB-induced endometrial cancer, compared to the standard paclitaxel.
Collapse
Affiliation(s)
- Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Rocklands, Ooty, Tamil Nadu, 643001, India
| | - Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Rocklands, Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Rocklands, Ooty, Tamil Nadu, 643001, India.
| |
Collapse
|
10
|
Davanzo GG, Castro G, Moraes-Vieira PMM. Immunometabolic regulation of adipose tissue resident immune cells. Curr Opin Pharmacol 2021; 58:44-51. [PMID: 33878567 DOI: 10.1016/j.coph.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022]
Abstract
Adipose tissue (AT) performs immunoregulatory functions beyond fat storage. In addition to adipocytes, AT has a diverse spectrum of resident and infiltrating immune cells in health and disease. Immune cells contribute to the homeostatic function of AT by adapting their metabolism in accordance with the microenvironment. However, how the metabolic reprogramming of immune cells affects their inflammatory profile and the subsequent implication for adipocyte function is not completely elucidated. Here, we discuss the available data on metabolic regulatory processes implicated in the control of adipose tissue-resident immune cells and their crosstalk with adipocytes.
Collapse
Affiliation(s)
- Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | - Gisele Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Pedro Manoel M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, SP, Brazil.
| |
Collapse
|
11
|
Mu YT, Feng HH, Yu JQ, Liu ZK, Wang Y, Shao J, Li RH, Li DK. Curcumin suppressed proliferation and migration of human retinoblastoma cells through modulating NF-κB pathway. Int Ophthalmol 2020; 40:2435-2440. [PMID: 32399774 DOI: 10.1007/s10792-020-01406-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/27/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE To study the effect of curcumin on proliferation and invasion of the human retinoblastoma cells and its potential mechanism. METHODS A cell line of retinoblastoma (WERI-Rb-1) was treated with various concentrations of curcumin (0-40 µM). Cell number was counted with CCK8 kit, and cell migration was assessed using the Transwell assay. Immunoblotting was performed to detect the proteins of metalloproteinase-2 (MMP-2), MMP-9 and vascular endothelial growth factor (VEGF) as well as nuclear translocation of nuclear factor-κB (NF-κB, p65). RESULTS Proliferation and migration of WERI-Rb-1 cells were significantly inhibited by curcumin in a concentration-dependent manner (0-40 µM). Protein expressions of MMP-2, MMP-9 and VEGF in the WERI-Rb-1 cells were also significantly inhibited by curcumin in a concentration-dependent manner (0-40 µM). Furthermore, nuclear translocation of NF-κB (p65) was significantly inhibited by curcumin in time-dependent manner (6-24 h). CONCLUSION Curcumin inhibited proliferation and migration of WERI-Rb-1 cells, a cell line of human retinoblastoma, which might be through modulating NF-κB and its downstream proteins including VEGF, MMP-2, and MMP-9.
Collapse
Affiliation(s)
- Ying-Tao Mu
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China
| | - Huan-Huan Feng
- Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Jin-Qiang Yu
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China
| | - Zhen-Kai Liu
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China
| | - Yan Wang
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China
| | - Jie Shao
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China
| | - Ran-Hui Li
- Institute of Pathogen Biology, University of South China, Heyang, Hunan, China
| | - De-Kun Li
- Department of Ophthalmology, Shiyan Renmin Hospital, Hubei University of Medicine, No.39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China.
| |
Collapse
|
12
|
Dandu K, Kallamadi PR, Thakur SS, Rao CM. Drug Repurposing for Retinoblastoma: Recent Advances. Curr Top Med Chem 2019; 19:1535-1544. [PMID: 30659544 DOI: 10.2174/1568026619666190119152706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
Retinoblastoma is the intraocular malignancy that occurs during early childhood. The current standard of care includes chemotherapy followed by focal consolidative therapies, and enucleation. Unfortunately, these are associated with many side and late effects. New drugs and/or drug combinations need to be developed for safe and effective treatment. This compelling need stimulated efforts to explore drug repurposing for retinoblastoma. While conventional drug development is a lengthy and expensive process, drug repurposing is a faster, alternate approach, where an existing drug, not meant for treating cancer, can be repurposed to treat retinoblastoma. The present article reviews various attempts to test drugs approved for different purposes such as calcium channels blockers, non-steroidal antiinflammatory drugs, cardenolides, antidiabetic, antibiotics and antimalarial for treating retinoblastoma. It also discusses other promising candidates that could be explored for repurposing for retinoblastoma.
Collapse
Affiliation(s)
- Kamakshi Dandu
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Habsiguda, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Prathap R Kallamadi
- School of Life Sciences. University of Hyderabad, Prof. C.R. Rao Road, Hyderabad 500 046, India
| | - Suman S Thakur
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Habsiguda, Hyderabad 500 007, India
| | - Ch Mohan Rao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Habsiguda, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India
| |
Collapse
|
13
|
Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur J Med Chem 2019; 166:502-513. [DOI: 10.1016/j.ejmech.2019.01.067] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
|
14
|
Kao LT, Xirasagar S, Lin HC, Huang CY. Association Between Pioglitazone Use and Prostate Cancer: A Population-Based Case-Control Study in the Han Population. J Clin Pharmacol 2018; 59:344-349. [DOI: 10.1002/jcph.1326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Li-Ting Kao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan.,Sleep Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sudha Xirasagar
- Department of Health Services Policy and Management, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Herng-Ching Lin
- Sleep Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,School of Health Care Administration, Taipei Medical University, Taipei, Taiwan
| | - Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, Yun-Lin Branch, Douliu, Taiwan.,Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taiwan.,School of Public Health, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
A Sterol from Soft Coral Induces Apoptosis and Autophagy in MCF-7 Breast Cancer Cells. Mar Drugs 2018; 16:md16070238. [PMID: 30018246 PMCID: PMC6071057 DOI: 10.3390/md16070238] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor that plays a key role in regulating cellular metabolism, and is a therapeutic target for cancer therapy. To search for potential PPARγ activators, a compound library comprising 11 marine compounds was examined. Among them, a sterol, 3β,11-dihydroxy-9,11-secogorgost-5-en-9-one (compound 1), showed the highest PPARγ activity with an IC50 value of 8.3 μM for inhibiting human breast adenocarcinoma cell (MCF-7) growth. Western blotting experiments showed that compound 1 induces caspase activation and PARP cleavage. In addition, compound 1 modulated the expression of various PPARγ-regulated downstream biomarkers including cyclin D1, cyclin-dependent kinase (CDK)6, B-cell lymphoma 2 (Bcl-2), p38, and extracellular-signal-regulated kinase (ERK). Moreover, compound 1 increased reactive oxygen species (ROS) generation, upregulated the phosphorylation and expression of H2AX, and induced autophagy. Interestingly, pre-treatment with the autophagy inhibitor 3-methyladenine rescued cells from compound 1-induced growth inhibition, which indicates that the cytotoxic effect of compound 1 is, in part, attributable to its ability to induce autophagy. In conclusion, these findings suggest the translational potential of compound 1 in breast cancer therapy.
Collapse
|
16
|
Human Skin Permeation Studies with PPARγ Agonist to Improve Its Permeability and Efficacy in Inflammatory Processes. Int J Mol Sci 2017; 18:ijms18122548. [PMID: 29182532 PMCID: PMC5751151 DOI: 10.3390/ijms18122548] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 01/09/2023] Open
Abstract
Rosacea is the most common inflammatory skin disease. It is characterized by erythema, inflammatory papules and pustules, visible blood vessels, and telangiectasia. The current treatment has limitations and unsatisfactory results. Pioglitazone (PGZ) is an agonist of peroxisome proliferator-activated receptors (PPARs), a nuclear receptor that regulates important cellular functions, including inflammatory responses. The purpose of this study was to evaluate the permeation of PGZ with a selection of penetration enhancers and to analyze its effectiveness for treating rosacea. The high-performance liquid chromatography (HPLC) method was validated for the quantitative determination of PGZ. Ex vivo permeation experiments were realized in Franz diffusion cells using human skin, in which PGZ with different penetration enhancers were assayed. The results showed that the limonene was the most effective penetration enhancer that promotes the permeation of PGZ through the skin. The cytotoxicity studies and the Draize test detected cell viability and the absence of skin irritation, respectively. The determination of the skin color using a skin colorimetric probe and the results of histopathological studies confirmed the ability of PGZ-limonene to reduce erythema and vasodilation. This study suggests new pharmacological indications of PGZ and its possible application in the treatment of skin diseases, namely rosacea.
Collapse
|
17
|
Epigallocatechin Gallate Attenuates β-Amyloid Generation and Oxidative Stress Involvement of PPARγ in N2a/APP695 Cells. Neurochem Res 2016; 42:468-480. [PMID: 27889855 DOI: 10.1007/s11064-016-2093-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/24/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
Abstract
The accumulation of β-amyloid (Aβ) peptide plaques is a major pathogenic event in Alzheimer's disease (AD). Aβ is a cleaved fragment of APP via BACE1, which is the rate-limiting enzyme in APP processing and Aβ generation. Nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is considered to be a potential target for AD treatment, because of its potent antioxidant and inhibitory effects on Aβ production by negatively regulating BACE1. Epigallocatechin gallate (EGCG), a highly active catechin found in green tea, is known to enhance metabolic activity and cognitive ability in the mice model of AD. To investigate whether the therapeutic effect of EGCG is related to the PPARγ pathway, we analysed the alterations in the intracellular molecular expression of PPARγ after EGCG treatment in the N2a/APP695 cell line. In this study, we observed that EGCG attenuated Aβ generation in N2a/APP695 cells, such as the PPARγ agonist, pioglitazone, by suppressing the transcription and translation of BACE1 and that its effect was attenuated by the PPARγ inhibitor, GW9662. Intriguingly, EGCG significantly reinforced the activity of PPARγ by promoting its mRNA and protein expressions in N2a/APP695 cells. Moreover, EGCG also decreased the expression of pro-apoptotic proteins (Bax, caspase-3), reduced the activity of the anti-inflammatory agent NF-κB and inhibited the oxidative stress by decreasing the levels of ROS and MDA and increasing the expression of MnSOD. Co-administration of GW9662 also significantly decreased the EGCG-mediated neuroprotective effect evidenced by the increase in oxidative stress and inflammatory markers. The therapeutic efficacy of EGCG in AD may be derived from the up-regulation of PPARγ mRNA and protein expressions.
Collapse
|