1
|
Mouse Models for Application in Colorectal Cancer: Understanding the Pathogenesis and Relevance to the Human Condition. Biomedicines 2022; 10:biomedicines10071710. [PMID: 35885015 PMCID: PMC9313309 DOI: 10.3390/biomedicines10071710] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer (CRC) is a malignant disease that is the second most common cancer worldwide. CRC arises from the complex interactions among a variety of genetic and environmental factors. To understand the mechanism of colon tumorigenesis, preclinical studies have developed various mouse models including carcinogen-induced and transgenic mice to recapitulate CRC in humans. Using these mouse models, scientific breakthroughs have been made on the understanding of the pathogenesis of this complex disease. Moreover, the availability of transgenic knock-in or knock-out mice further increases the potential of CRC mouse models. In this review, the overall features of carcinogen-induced (focusing on azoxymethane and azoxymethane/dextran sulfate sodium) and transgenic (focusing on ApcMin/+) mouse models, as well as their mechanisms to induce colon tumorigenesis, are explored. We also discuss limitations of these mouse models and their applications in the evaluation and study of drugs and treatment regimens against CRC. Through these mouse models, a better understanding of colon tumorigenesis can be achieved, thereby facilitating the discovery of novel therapeutic strategies against CRC.
Collapse
|
2
|
Khafaga AF, Shamma RN, Abdeen A, Barakat AM, Noreldin AE, Elzoghby AO, Sallam MA. Celecoxib repurposing in cancer therapy: molecular mechanisms and nanomedicine-based delivery technologies. Nanomedicine (Lond) 2021; 16:1691-1712. [PMID: 34264123 DOI: 10.2217/nnm-2021-0086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While cancer remains a significant global health problem, advances in cancer biology, deep understanding of its underlaying mechanism and identification of specific molecular targets allowed the development of new therapeutic options. Drug repurposing poses several advantages as reduced cost and better safety compared with new compounds development. COX-2 inhibitors are one of the most promising drug classes for repurposing in cancer therapy. In this review, we provide an overview of the detailed mechanism and rationale of COX-2 inhibitors as anticancer agents and we highlight the most promising research efforts on nanotechnological approaches to enhance COX-2 inhibitors delivery with special focus on celecoxib as the most widely studied agent for chemoprevention or combined with chemotherapeutic and herbal drugs for combating various cancers.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, 13736, Egypt
| | | | - Ahmed E Noreldin
- Department of Histology & Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22516, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Marwa A Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
3
|
Khodaverdi E, Eisvand F, Nezami MS, Shiadeh SNR, Kamali H, Hadizadeh F. Injectable In-Situ Forming Depot of Doxycycline Hyclate/α-Cyclodextrin Complex Using PLGA for Periodontitis Treatment: Preparation, Characterization, and In-Vitro Evaluation. Curr Drug Deliv 2020; 18:729-740. [PMID: 33155908 DOI: 10.2174/1567201817999201103195104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/29/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Doxycycline (DOX) is used in treating a bacterial infection, especially for periodontitis treatment. OBJECTIVE To reduce irritation of DOX for subgingival administration and increase the chemical stability and against enzymatic, the complex of α-cyclodextrin with DOX was prepared and loaded into injectable in situ forming implant based on PLGA. METHODS FTIR, molecular docking studies, X-ray diffraction, and differential scanning calorimetry was performed to characterize the DOX/α-cyclodextrin complex. Finally, the in-vitro drug release and modeling, morphological properties, and cellular cytotoxic effects were also evaluated. RESULTS The stability of DOX was improved with complex than pure DOX. The main advantage of the complex is the almost complete release (96.31 ± 2.56 %) of the drug within 14 days of the implant, whereas in the formulation containing the pure DOX and the physical mixture the DOX with α-cyclodextrin release is reached to 70.18 ± 3.61 % and 77.03 ± 3.56 %, respectively. This trend is due to elevate of DOX stability in the DOX/ α-cyclodextrin complex form within PLGA implant that confirmed by the results of stability. CONCLUSION Our results were indicative that the formulation containing DOX/α-cyclodextrin complex was biocompatible and sustained-release with minimum initial burst release.
Collapse
Affiliation(s)
- Elham Khodaverdi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Eisvand
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Sina Nezami
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nesa Rezaeian Shiadeh
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Çoban Ö, Değim Z, Yılmaz Ş, Altıntaş L, Arsoy T, Sözmen M. Efficacy of targeted liposomes and nanocochleates containing imatinib plus dexketoprofen against fibrosarcoma. Drug Dev Res 2019; 80:556-565. [PMID: 30901500 DOI: 10.1002/ddr.21530] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/27/2019] [Accepted: 03/03/2019] [Indexed: 11/06/2022]
Abstract
The main challenges in treating cancer using chemotherapeutics are insufficient dose at the target site and the development of drug resistance, while higher doses can induce side effects by damaging nontarget tissues. Combinatorial drug therapy may overcome these limitations by permitting lower doses and more specific targeting, thereby mitigating drug resistance and nontarget side effects. Recent reports indicate that nonsteroidal anti-inflammatory drugs (NSAIDs) have anticancer potential and can be used together with conventional chemotherapeutics to improve efficacy and safety. In the present study, imatinib mesylate and dexketoprofen trometamol were selected as model drugs to develop targeted surface-modified liposome and nanocochleate formulations for fibrosarcoma treatment. The physicochemical properties and in vitro efficacy of various formulations were evaluated by measurement of particle size distribution, polydispersity index, zeta potential, encapsulation efficiency, diffusion through Caco-2 cells, and toxicity in culture. Selected formulations were then evaluated in fibrosarcoma-bearing model mice by histopathological observations and tyrosine kinase receptor inhibition assays. The most effective formulation on the fibrosarcoma model was a PEGylated nanocochleate formulation. These findings provide a foundation for developing more effective formulations and chemotherapeutic strategies for the treatment of fibrosarcoma and other types of cancer.
Collapse
Affiliation(s)
- Özlem Çoban
- Department of Pharmaceutical Technology, Karadeniz Technical University Faculty of Pharmacy, Trabzon, Turkey
| | - Zelihagül Değim
- Department of Pharmaceutical Technology, Biruni University Faculty of Pharmacy, İstanbul, Turkey
| | - Şükran Yılmaz
- Department of Cell Bank, Food and Mouth Diseases Institute, Ankara, Turkey
| | - Levent Altıntaş
- Department of Pharmacology and Toxicology, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| | - Taibe Arsoy
- Department of Cell Bank, Food and Mouth Diseases Institute, Ankara, Turkey
| | - Mahmut Sözmen
- Department of Preclinical Sciences, OndokuzMayıs University, Faculty of Veterinary Medicine, Samsun, Turkey
| |
Collapse
|
5
|
Nasr T, Bondock S, Rashed HM, Fayad W, Youns M, Sakr TM. Novel hydrazide-hydrazone and amide substituted coumarin derivatives: Synthesis, cytotoxicity screening, microarray, radiolabeling and in vivo pharmacokinetic studies. Eur J Med Chem 2018; 151:723-739. [PMID: 29665526 DOI: 10.1016/j.ejmech.2018.04.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/31/2018] [Accepted: 04/06/2018] [Indexed: 01/04/2023]
Abstract
The current work presents the synthesis and biological evaluation of new series of coumarin hydrazide-hydrazone derivatives that showed in vitro broad spectrum antitumor activities against resistant pancreatic carcinoma (Panc-1), hepatocellular carcinoma (HepG2) and leukemia (CCRF) cell lines using doxorubicin as reference standard. Bromocoumarin hydrazide-hydrazone derivative (BCHHD) 11b showed excellent anticancer activity against all tested cancer cell lines. Enzyme assays showed that BCHHD 11b induced apoptosis due to activation of caspases 3/7. Moreover, 11b inhibited GST and CYP3A4 in a dose dependent manner and the induced cell death could be attributed to metabolic inhibition. Moreover, 11b microarray analysis showed significant up- and down-regulation of many genes in the treated cells related to apoptosis, cell cycle, tumor growth and suppressor genes. All of the above presents BCHHD 11b as a potent anticancer agent able to overcome drug resistance. In addition, compound 11b was able to serve as a chemical carrier for 99mTc and the in vivo biodistribution study of 99mTc-11b complex revealed a remarkable targeting ability of 99mTc into solid tumor showing that 99mTc-11b might be used as a promising radiopharmaceutical imaging agent for cancer.
Collapse
Affiliation(s)
- Tamer Nasr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Modern University for Technology and Information, Egypt.
| | - Samir Bondock
- Department of Chemistry, Faculty of Science, Mansoura University, ET-35516, Mansoura, Egypt; Department of Chemistry, Faculty of Science, King Khalid University, 9004, Abha, Saudi Arabia
| | - Hassan M Rashed
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, P.O. Code 13759, Cairo, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Mahmoud Youns
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Egypt.
| | - Tamer M Sakr
- Radioactive Isotopes and Generators Department, Hot Laboratories Centre, Atomic Energy Authority, P.O. Code 13759, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, October University of Modern Sciences and Arts (MSA), Giza, Egypt.
| |
Collapse
|
6
|
Tsai CC, Li YS, Lin PP. Inonotus obliquus extract induces apoptosis in the human colorectal carcinoma’s HCT-116 cell line. Biomed Pharmacother 2017; 96:1119-1126. [DOI: 10.1016/j.biopha.2017.11.111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/19/2017] [Accepted: 11/20/2017] [Indexed: 01/01/2023] Open
|
7
|
Hashemzaei M, Delarami Far A, Yari A, Heravi RE, Tabrizian K, Taghdisi SM, Sadegh SE, Tsarouhas K, Kouretas D, Tzanakakis G, Nikitovic D, Anisimov NY, Spandidos DA, Tsatsakis AM, Rezaee R. Anticancer and apoptosis‑inducing effects of quercetin in vitro and in vivo. Oncol Rep 2017; 38:819-828. [PMID: 28677813 PMCID: PMC5561933 DOI: 10.3892/or.2017.5766] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/13/2017] [Indexed: 12/16/2022] Open
Abstract
The present study focused on the elucidation of the putative anticancer potential of quercetin. The anticancer activity of quercetin at 10, 20, 40, 80 and 120 µM was assessed in vitro by MMT assay in 9 tumor cell lines (colon carcinoma CT‑26 cells, prostate adenocarcinoma LNCaP cells, human prostate PC3 cells, pheocromocytoma PC12 cells, estrogen receptor‑positive breast cancer MCF‑7 cells, acute lymphoblastic leukemia MOLT‑4 T‑cells, human myeloma U266B1 cells, human lymphoid Raji cells and ovarian cancer CHO cells). Quercetin was found to induce the apoptosis of all the tested cancer cell lines at the utilized concentrations. Moreover, quercetin significantly induced the apoptosis of the CT‑26, LNCaP, MOLT‑4 and Raji cell lines, as compared to control group (P<0.001), as demonstrated by Annexin V/PI staining. In in vivo experiments, mice bearing MCF‑7 and CT‑26 tumors exhibited a significant reduction in tumor volume in the quercetin‑treated group as compared to the control group (P<0.001). Taken together, quercetin, a naturally occurring compound, exhibits anticancer properties both in vivo and in vitro.
Collapse
Affiliation(s)
- Mahmoud Hashemzaei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Amin Delarami Far
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Arezoo Yari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Reza Entezari Heravi
- Students Research Committee, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Kaveh Tabrizian
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sarvenaz Ekhtiari Sadegh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | | | - Dimitrios Kouretas
- Department of Biochemistry and Biotechnology, Faculty of Animal Physiology‑Toxicology, University of Thessaly, Larissa, Greece
| | - George Tzanakakis
- Department of Anatomy‑Histology‑Embryology, Medical School, University of Crete, Greece
| | - Dragana Nikitovic
- Department of Anatomy‑Histology‑Embryology, Medical School, University of Crete, Greece
| | | | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Aristides M Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Jin GH, Xu W, Shi Y, Wang LB. Celecoxib exhibits an anti-gastric cancer effect by targeting focal adhesion and leukocyte transendothelial migration-associated genes. Oncol Lett 2016; 12:2345-2350. [PMID: 27698798 PMCID: PMC5038604 DOI: 10.3892/ol.2016.4976] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer (GC) is a prevalent cancer, which remains incurable, and therefore requires an alternative treatment method. Celecoxib is a nonsteroidal anti-inflammatory drug that targets cyclooxygenase-2, and exhibits anticancer effects. The present study aimed to investigate the anti-GC mechanism of celecoxib using bioinformatics methods. Gene expression datasets GSE56807 (GC tissues and normal gastric tissues) and GSE54657 (celecoxib-treated and non-treated human GC epithelial AGS cells) were downloaded from the Gene Expression Omnibus database. Two groups of differentially expressed genes (DEGs) were identified using limma package in R language. The criterion for GSE56807 was a false discovery rate of <0.05, while that for GSE54657 was P<0.01. Overlapping DEGs from the two datasets were screened out. Subsequently, pathway enrichment analysis was performed using Database for Annotation, Visualization and Integrated Discovery software (P<0.1; gene count ≥2). In addition, the protein-protein interactions (PPIs) among the overlapped DEGs were obtained based on IntAct, Database of Interacting Proteins, Biomolecular Interaction Network Database and Human Protein Reference Database. Finally, a PPI network was visualized using Cytoscape software. A total of 137 overlapped DEGs were obtained, and DEGs with opposite regulation directions in the two datasets were significantly enriched in focal adhesion and leukocyte transendothelial migration. Subsequently, a PPI network of overlapped DEGs was constructed. Comprehensively, a total of 8 key DEGs [cysteine and glycine rich protein 1 (CSRP1), thrombospondin 1 (THBS1), myosin light chain 9 (MYL9), filamin A (FLNA), actinin alpha 1 (ACTN1), vinculin (VCL), laminin subunit gamma 2 (LAMC2) and claudin 1 (CLDN1)] were upregulated in GC tissues and downregulated in celecoxib-treated cells. In conclusion, celecoxib may exhibit anti-GC effects by suppressing the expression of CSRP1, THBS1, MYL9, FLNA, ACTN1, VCL, LAMC2 and CLDN1, and inhibiting leukocyte transendothelial migration and focal adhesion. However, relevant experiments are required to confirm the conclusion of the present study.
Collapse
Affiliation(s)
- Guo-Hua Jin
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Xu
- Department of Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Shi
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li-Bo Wang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
9
|
Jang Y, Park NY, Rostgaard-Hansen AL, Huang J, Jiang Q. Vitamin E metabolite 13'-carboxychromanols inhibit pro-inflammatory enzymes, induce apoptosis and autophagy in human cancer cells by modulating sphingolipids and suppress colon tumor development in mice. Free Radic Biol Med 2016; 95:190-9. [PMID: 27016075 PMCID: PMC4867259 DOI: 10.1016/j.freeradbiomed.2016.03.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/29/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023]
Abstract
Vitamin E forms are substantially metabolized to various carboxychromanols including 13'-carboxychromanols (13'-COOHs) that are found at high levels in feces. However, there is limited knowledge about functions of these metabolites. Here we studied δT-13'-COOH and δTE-13'-COOH, which are metabolites of δ-tocopherol and δ-tocotrienol, respectively. δTE-13'-COOH is also a natural constituent of a traditional medicine Garcinia Kola. Both 13'-COOHs are much stronger than tocopherols in inhibition of pro-inflammatory and cancer promoting cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX), and in induction of apoptosis and autophagy in colon cancer cells. The anticancer effects by 13'-COOHs appeared to be partially independent of inhibition of COX-2/5-LOX. Using liquid chromatography tandem mass spectrometry, we found that 13'-COOHs increased intracellular dihydrosphingosine and dihydroceramides after short-time incubation in HCT-116 cells, and enhanced ceramides while decreased sphingomyelins during prolonged treatment. Modulation of sphingolipids by 13'-COOHs was observed prior to or coinciding with biochemical manifestation of cell death. Pharmaceutically blocking the increase of these sphingolipids partially counteracted 13'-COOH-induced cell death. Further, 13'-COOH inhibited dihydroceramide desaturase without affecting the protein expression. In agreement with these mechanistic findings, δTE-13'-COOH significantly suppressed the growth and multiplicity of colon tumor in mice. Our study demonstrates that 13'-COOHs have anti-inflammatory and anticancer activities, may contribute to in vivo anticancer effect of vitamin E forms and are promising novel cancer prevention agents.
Collapse
Affiliation(s)
- Yumi Jang
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Na-Young Park
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | | | - Jianjie Huang
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Qing Jiang
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
10
|
Synthesis, biological activity screening and molecular modeling study of acylaminoacetamide derivatives. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1419-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
11
|
Niu X, Mu Q, Li W, Yao H, Li H, Li Y, Hu H, Huang H. Protective effects of esculentic acid against endotoxic shock in Kunming mice. Int Immunopharmacol 2014; 23:229-35. [PMID: 25242384 DOI: 10.1016/j.intimp.2014.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/12/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
Abstract
Esculentic acid (EA), a triterpene compound extracted from the root of Phytolacca esculenta (the Chinese name Shang Lu), has been widely used to therapy a variety of inflammatory diseases such as rheumatoid arthritis, edema, hepatitis and bronchitis. The present study was designed to investigate the protective effects of EA against LPS-induced endotoxic shock by the intraperitoneal injection of EA (1, 5 and 10 mg/kg) prior to LPS stimulation (1 mg/kg, i.p.). We examined the effects of EA on the survival rate of mice, inflammatory cytokine and pro-inflammatory mediator production, histopathological changes and protein expression of COX-2 in tissue sections from lung, liver and kidney. The results indicate that EA not only increases the survival rate of mice, but decreases the levels of TNF-α, IL-6, NO and PGE2 in serum or tissues, histopathological changes and COX-2 protein expression also. Furthermore, EA also increases the levels of anti-inflammatory cytokine IL-10 in serum. Overall, these data suggest that the protective effects of EA against LPS-induced endotoxic shock may be mediated, at least in part, by regulation the release of inflammatory cytokines and mediators, and protein expression of COX-2 in mice.
Collapse
Affiliation(s)
- Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China.
| | - Qingli Mu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China.
| | - Huan Yao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Huani Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Yongmei Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Hua Hu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Huimin Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
12
|
Jang JE, Kim HM, Kim H, Jeon DY, Park CH, Kwon SY, Chung JW, Khang G. Inflammatory Responses to Hydroxyapatite/Poly(lactic-co-glycolic acid) Scaffolds with Variation of Compositions. POLYMER-KOREA 2014. [DOI: 10.7317/pk.2014.38.2.156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Wang YJ, Niu XP, Yang L, Han Z, Ma YJ. Effects of celecoxib on cycle kinetics of gastric cancer cells and protein expression of cytochrome C and caspase-9. Asian Pac J Cancer Prev 2013; 14:2343-7. [PMID: 23725138 DOI: 10.7314/apjcp.2013.14.4.2343] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE This investigation aimed to determine effects of celecoxib on the cell cycle kinetics of the gastric cancer cell line MGC803 and the mechanisms involved by assessing expression of cytochrome C and caspase-9 at the protein level. METHODS Cell proliferation of MGC803 was determined by MTT assay after treatment with celecoxib. Apoptosis was assessed using fluorescence staining and cell cycle kinetics by flow cytometry. Western blotting was used to detect the expression of caspase-9 protein and of cytochrome C protein in cell cytosol and mitochondria. RESULTS Celecoxib was able to restrain proliferation and induce apoptosis in a dose- and time- dependent manner, inducing G0/G1 cell cycle arrest, release of cytochrome C into the cytosol, and cleavage of pro-caspase-9 into its active form. CONCLUSION Celecoxib can induce apoptosis in MGC803 cells through a mechanism involving cell cycle arrest, mitochondrial cytochrome C release and caspase activation.
Collapse
Affiliation(s)
- Yu-Jie Wang
- Department of Gastroenterology, Zhengzhou People's Hospital Affiliated to Southern Medical University, Zhengzhou, Henan, China
| | | | | | | | | |
Collapse
|
14
|
Sharma R, Rawal RK, Gaba T, Singla N, Malhotra M, Matharoo S, Bhardwaj TR. Design, synthesis and ex vivo evaluation of colon-specific azo based prodrugs of anticancer agents. Bioorg Med Chem Lett 2013; 23:5332-8. [PMID: 23968824 DOI: 10.1016/j.bmcl.2013.07.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/18/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
Colon-specific azo based prodrugs of anticancer agents like methotrexate (6), gemcitabine (7) and analogue of oxaliplatin (RTB-4) (8) were synthesized and characterized by modern analytical techniques. The prepared prodrugs were stable in acidic (pH 1.2) and basic (pH 7.4) buffers which showed their stability in upper GIT environment. Further, an assay was performed which demonstrated the presence of azoreductase enzyme in the rat fecal material, rat cecum content and other parts of intestinal content which reduce specifically the azo bond and release the drug. The in vitro cytotoxicity assay was also performed which clearly indicated that these azo based prodrugs are active against human colorectal cancer cell lines (COLO 205, COLO 320 DM and HT-29). The release behavior of prodrugs (10, 11 and 15) was 60-70% after 24h incubation at 37°C. Therefore, the synthesized azo linked prodrugs of methotrexate, gemcitabine and RTB-4 are the potential candidates for colon targeted drug delivery system with minimal undesirable side effects.
Collapse
Affiliation(s)
- Rajiv Sharma
- Department of Pharmaceutical Chemistry, Indo-Soviet Friendship (ISF) College of Pharmacy, Moga, India;
| | | | | | | | | | | | | |
Collapse
|
15
|
Tao YF, Lu J, Du XJ, Sun LC, Zhao X, Peng L, Cao L, Xiao PF, Pang L, Wu D, Wang N, Feng X, Li YH, Ni J, Wang J, Pan J. Survivin selective inhibitor YM155 induce apoptosis in SK-NEP-1 Wilms tumor cells. BMC Cancer 2012; 12:619. [PMID: 23267699 PMCID: PMC3543843 DOI: 10.1186/1471-2407-12-619] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 12/21/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Survivin, a member of the family of inhibitor of apoptosis proteins, functions as a key regulator of mitosis and programmed cell death. YM155, a novel molecular targeted agent, suppresses survivin, which is overexpressed in many tumor types. The aim of this study was to determine the antitumor activity of YM155 in SK-NEP-1 cells. METHODS SK-NEP-1 cell growth in vitro and in vivo was assessed by MTT and nude mice experiments. Annexin V/propidium iodide staining followed by flow cytometric analysis was used to detect apoptosis in cell culture. Then gene expression profile of tumor cells treated with YM155 was analyzed with real-time PCR arrays. We then analyzed the expression data with MEV (Multi Experiment View) cluster software. Datasets representing genes with altered expression profile derived from cluster analyses were imported into the Ingenuity Pathway Analysis tool. RESULTS YM155 treatment resulted in inhibition of cell proliferation of SK-NEP-1cells in a dose-dependent manner. Annexin V assay, cell cycle, and activation of caspase-3 demonstrates that YM155 induced apoptosis in SK-NEP-1 cells. YM155 significantly inhibited growth of SK-NEP-1 xenografts (YM155 5 mg/kg: 1.45 ± 0.77 cm3; YM155 10 mg/kg: 0.95 ± 0.55 cm3) compared to DMSO group (DMSO: 3.70 ± 2.4 cm3) or PBS group cells (PBS: 3.78 ± 2.20 cm3, ANOVA P < 0.01). YM155 treatment decreased weight of tumors (YM155 5 mg/kg: 1.05 ± 0.24 g; YM155 10 mg/kg: 0.72 ± 0.17 g) compared to DMSO group (DMSO: 2.06 ± 0.38 g) or PBS group cells (PBS: 2.36 ± 0.43 g, ANOVA P < 0.01). Real-time PCR array analysis showed between Test group and control group there are 32 genes significantly up-regulated and 54 genes were significantly down-regulated after YM155 treatment. Ingenuity pathway analysis (IPA) showed cell death was the highest rated network with 65 focus molecules and the significance score of 44. The IPA analysis also groups the differentially expressed genes into biological mechanisms that are related to cell death, cellular function maintenance, cell morphology, carbohydrate metabolism and cellular growth and proliferation. Death receptor signaling (3.87E-19), TNFR1 signaling, induction of apoptosis by HIV1, apoptosis signaling and molecular mechanisms of cancer came out to be the top four most significant pathways. IPA analysis also showed top molecules up-regulated were BBC3, BIRC3, BIRC8, BNIP1, CASP7, CASP9, CD5, CDKN1A, CEBPG and COL4A3, top molecules down-regulated were ZNF443, UTP11L, TP73, TNFSF10, TNFRSF1B, TNFRSF25, TIAF1, STK17A, SST and SPP1, upstream regulator were NR3C1, TP53, dexamethasone , TNF and Akt. CONCLUSIONS The present study demonstrates that YM155 treatment resulted in apoptosis and inhibition of cell proliferation of SK-NEP-1cells. YM155 had significant role and little side effect in the treatment of SK-NEP-1 xenograft tumors. Real-time PCR array analysis firstly showed expression profile of genes dyes-regulated after YM155 treatment. IPA analysis also represents new molecule mechanism of YM155 treatment, such as NR3C1 and dexamethasone may be new target of YM155. And our results may provide new clues of molecular mechanism of apoptosis induced by YM155.
Collapse
Affiliation(s)
- Yan-Fang Tao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Xiao-Juan Du
- Department of Gastroenterology, the 5th Hospital of Chinese PLA, Yin chuan, Ningxia Province, China
| | - Li-Chao Sun
- Department of Cell and Molecular Biology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xuan Zhao
- Beijing Insititute for Drug Control, Beijing, China
| | - Liang Peng
- Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Lan Cao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Pei-Fang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Li Pang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Dong Wu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Na Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Xing Feng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Yan-Hong Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Ni
- Translational Research Center, Second Hospital, The Second Clinical School, Nanjing Medical University, Nanjing, China
| | - Jian Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Heravi RE, Hadizadeh F, Sankian M, Afshari JT, Behravan J. Cyclooxygenase-2 inhibition by novel Bisaryl imidazolyl imidazole derivatives increases Bax/Bcl-2 ratio and upregulates Caspase-3 gene expression in Caco-2 colorectal cancer cell line. Genes Genomics 2012. [DOI: 10.1007/s13258-011-0168-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|