1
|
Gao J, Song G, Shen H, Wu Y, Zhao C, Zhang Z, Jiang Q, Li X, Ma X, Tan B, Yin Y. Allicin Improves Intestinal Epithelial Barrier Function and Prevents LPS-Induced Barrier Damages of Intestinal Epithelial Cell Monolayers. Front Immunol 2022; 13:847861. [PMID: 35185936 PMCID: PMC8854216 DOI: 10.3389/fimmu.2022.847861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
Gut barrier disruption is the initial pathogenesis of various diseases. We previously reported that dietary allicin improves tight junction proteins in the endoplasmic reticulum stressed jejunum. However, whether the allicin benefits the gut barrier within mycotoxin or endotoxin exposure is unknown. In the present study, IPEC-J2 cell monolayers within or without deoxynivalenol (DON) or lipopolysaccharide (LPS) challenges were employed to investigate the effects of allicin on intestinal barrier function and explore the potential mechanisms. Results clarified that allicin at 2 μg/mL increased the viability, whereas the allicin higher than 10 μg/mL lowered the viability of IPEC-J2 cells via inhibiting cell proliferation. Besides, allicin increased trans-epithelial electric resistance (TEER), decreased paracellular permeability, and enhanced ZO-1 integrity of the IPEC-J2 cell monolayers. Finally, allicin supplementation prevented the LPS-induced barrier damages via activating Nrf2/HO-1 pathway-dependent antioxidant system. In conclusion, the present study strongly confirmed allicin as an effective nutrient to improve intestinal barrier function and prevent bacterial endotoxin-induced barrier damages.
Collapse
Affiliation(s)
- Jingxia Gao
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Guanzhong Song
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Haibo Shen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yiming Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chongqi Zhao
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhuo Zhang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Qian Jiang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Qian Jiang,
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaokang Ma
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Bie Tan
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yulong Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
2
|
Zhao B, Fan Y, Li H, Zhang C, Han R, Che D. Mitigative effects of Eleutheroside E against the mechanical barrier dysfunction induced by soybean agglutinin in IPEC-J2 cell line. J Anim Physiol Anim Nutr (Berl) 2022; 106:664-670. [PMID: 35014099 DOI: 10.1111/jpn.13677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/06/2021] [Accepted: 11/14/2021] [Indexed: 01/20/2023]
Abstract
Soybean agglutinin (SBA) is an anti-nutritional factor which decreases the mechanical barrier function in intestinal porcine jejunum epithelial cells (IPEC-J2). Eleutheroside E (EE) is a key part of Acanthopanax senticosus to exert pharmacological effects. This study aims to investigate the effects of EE on the barrier function in IPEC-J2 cells and to determine the ability of EE to enhance the protective effect of barrier function against SBA exposure. The IPEC-J2 cells were cultured in mediums with concentration of 0.1 mg/ml EE, 0.5 ml/ml SBA and 0.1 mg/ml EE pre-treated then treated with 0.5 mg/ml SBA. Then, the transepithelial electric resistance (TEER) value, inflammatory cytokines mRNA expression, tight junction mRNA and protein expression were tested by epithelial Voltohm meter, q-PCR and Western blot method respectively. The results showed that cells treated with 0.1 mg/ml EE had lower permeability (p < 0.05) while 0.5 mg/ml SBA treatment had higher permeability through tested TEER, and higher tight junction proteins (Claudin-3 and ZO-1) expressions and genes (Claudin-3, Occludin and ZO-1) expressions (p < 0.05) in 0.1 mg/ml EE group. IPEC-J2 cells pre-treated with 0.1 mg/ml EE could significantly improve the inflammatory response caused by 0.5 mg/ml SBA by up-regulation for IL-10, TGF-β, and down-regulation gene expression of IL-6, TNF-α and IFN-γ (p < 0.05). In conclusion, 0.1 mg/ml EE can improve the mechanical barrier function and could protect the effects while 0.5 mg/ml of SBA-induced barrier dysfunction in IPEC-J2.
Collapse
Affiliation(s)
- Bao Zhao
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Key Lab of Animal Production & Product Quality, and Security, Ministry of Education, Jilin Provincial Swine Industry Technical Innovation Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yueli Fan
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Key Lab of Animal Production & Product Quality, and Security, Ministry of Education, Jilin Provincial Swine Industry Technical Innovation Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Huijuan Li
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Key Lab of Animal Production & Product Quality, and Security, Ministry of Education, Jilin Provincial Swine Industry Technical Innovation Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chun Zhang
- College of Animal Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Rui Han
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Key Lab of Animal Production & Product Quality, and Security, Ministry of Education, Jilin Provincial Swine Industry Technical Innovation Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Dongsheng Che
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Key Lab of Animal Production & Product Quality, and Security, Ministry of Education, Jilin Provincial Swine Industry Technical Innovation Center, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
3
|
Wu J, He C, Bu J, Luo Y, Yang S, Ye C, Yu S, He B, Yin Y, Yang X. Betaine attenuates LPS-induced downregulation of Occludin and Claudin-1 and restores intestinal barrier function. BMC Vet Res 2020; 16:75. [PMID: 32131830 PMCID: PMC7057534 DOI: 10.1186/s12917-020-02298-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 02/21/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The intestinal epithelial barrier, which works as the first line of defense between the luminal environment and the host, once destroyed, it will cause serious inflammation or other intestinal diseases. Tight junctions (TJs) play a vital role to maintain the integrity of the epithelial barrier. Lipopolysaccharide (LPS), one of the most important inflammatory factors will downregulate specific TJ proteins including Occludin and Claudin-1 and impair integrity of the epithelial barrier. Betaine has excellent anti-inflammatory activity but whether betaine has any effect on TJ proteins, particularly on LPS-induced dysfunction of epithelial barriers remains unknown. The purpose of this study is to explore the pharmacological effect of betaine on improving intestinal barrier function represented by TJ proteins. Intestinal porcine epithelial cells (IPEC-J2) were used as an in vitro model. RESULTS The results demonstrated that betaine enhanced the expression of TJ proteins while LPS (1 μg/mL) downregulates the expression of these proteins. Furthermore, betaine attenuates LPS-induced decreases of TJ proteins both shown by Western blot (WB) and Reverse transcription-polymerase chain reaction (RT-PCR). The immunofluorescent images consistently revealed that LPS induced the disruption of TJ protein Claudin-1 and reduced its expression while betaine could reverse these alterations. Similar protective role of betaine on intestinal barrier function was observed by transepithelial electrical resistance (TEER) approach. CONCLUSION In conclusion, our research demonstrated that betaine attenuated LPS-induced downregulation of Occludin and Claudin-1 and restored the intestinal barrier function.
Collapse
Affiliation(s)
- Jingtao Wu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Caimei He
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Jie Bu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Yue Luo
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Shuyuan Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Chengyan Ye
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Silei Yu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China
| | - Binsheng He
- Changsha Medical University, Changsha, 410219, Hunan, People's Republic of China
- Institute of Subtropical Agriculture, Chinese Academy of Science, Changsha, 410125, People's Republic of China
| | - Yulong Yin
- Changsha Medical University, Changsha, 410219, Hunan, People's Republic of China
- Institute of Subtropical Agriculture, Chinese Academy of Science, Changsha, 410125, People's Republic of China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, 410013, People's Republic of China.
- Changsha Medical University, Changsha, 410219, Hunan, People's Republic of China.
- Institute of Subtropical Agriculture, Chinese Academy of Science, Changsha, 410125, People's Republic of China.
| |
Collapse
|
4
|
He C, Deng J, Hu X, Zhou S, Wu J, Xiao D, Darko KO, Huang Y, Tao T, Peng M, Wang Z, Yang X. Vitamin A inhibits the action of LPS on the intestinal epithelial barrier function and tight junction proteins. Food Funct 2019; 10:1235-1242. [PMID: 30747184 DOI: 10.1039/c8fo01123k] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammation caused by either intrinsic or extrinsic toxins results in intestinal barrier dysfunction, contributing to inflammatory bowel disease (IBD) and other diseases. Vitamin A is a widely used food supplement although its mechanistic effect on intestinal structures is largely unknown. The goal of this study was to explore the mechanism by investigating the influence of vitamin A on the intestinal barrier function, represented by tight junctions. IPEC-J2 cells were differentiated on transwell inserts and used as a model of intestinal barrier permeability. Transepithelial electrical resistance (TEER) was used as an indicator of monolayer integrity and paracellular permeability. Western blot and the reverse transcriptase-polymerase chain reaction were used to assess the protein and mRNA expression of tight junction proteins. Immunofluorescence microscopy was used to evaluate the localization and expression of tight junctions. Differentiated cells were treated with a vehicle control (Ctrl), inflammatory stimulus (1 μg mL-1 LPS), LPS co-treatment with 0.1 μmol L-1 Vitamin A (1 μg mL-1 LPS + 0.1 μmol L-1 VA) and 0.1 μmol L-1 Vitamin A. LPS significantly decreased TEER by 24 hours, continuing this effect to 48 hours after application. Vitamin A alleviated the LPS-induced decrease of TEER from 12 hours to 48 hours, while Vitamin A alone enhanced TEER, indicating that Vitamin A attenuated LPS-induced intestinal epithelium permeability. Mechanistically, different concentrations of Vitamin A (0-20 μmol L-1) enhanced tight junction protein markers including Zo-1, Occludin and Claudin-1 both at protein and mRNA levels with an optimized dose of 0.1 μmol L-1. Immunofluorescence results demonstrated that majority of Zo-1 and Claudin-1 is located at the tight junctions, as we expected. LPS reduced the expression of these proteins and Vitamin A reversed LPS-reduced expression of these proteins, consistent with the results of western blot. In conclusion, Vitamin A improves the intestinal barrier function and reverses LPS-induced intestinal barrier damage via enhancing the expression of tight junction proteins.
Collapse
Affiliation(s)
- Caimei He
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Che D, Zhao B, Fan Y, Han R, Zhang C, Qin G, Adams S, Jiang H. Eleutheroside B increase tight junction proteins and anti-inflammatory cytokines expression in intestinal porcine jejunum epithelial cells (IPEC-J2). J Anim Physiol Anim Nutr (Berl) 2019; 103:1174-1184. [PMID: 30990939 DOI: 10.1111/jpn.13087] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/11/2019] [Accepted: 02/20/2019] [Indexed: 12/23/2022]
Abstract
Eleutheroside B (EB) is a phenylpropanoid glycoside with anti-inflammatory properties, neuroprotective abilities, immunomodulatory effects, antinociceptive effects, and regulation of blood glucose. The aim of this study was to investigate the effects of EB on the barrier function in the intestinal porcine epithelial cells J2 (IPEC-J2). The IPEC-J2 cells were inoculated into 96-well plates at a density of 5 × 103 cells per well for 100% confluence. The cells were cultured in the presence of EB at concentrations of 0, 0.05, 0.10, and 0.20 mg/ml for 48 hr. Then, 0.10 mg/ml was selected as the suitable concentration for the estimation of transepithelial electric resistance (TEER) value, alkaline phosphatase activity, proinflammatory cytokines mRNA expression, tight junction mRNA and protein expression. The results of this study indicated that the supplementation of EB in IPEC-J2 cells decreased cellular membrane permeability and mRNA expression of proinflammatory cytokines, including interleukin-6 (IL-6), interferon-γ (INF-γ), and tumour necrosis factor-α (TNF-α). The supplementation of EB in IPEC-J2 cells increased tight junction protein expression and anti-inflammatory cytokines, interleukin 10 (IL-10) and transforming growth factor beta (TGF-β). In addition, the western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) results indicated that EB significantly (p < 0.05) increased the mRNA and protein expression of intestinal tight junction proteins, Claudin-3, Occludin, and Zonula Occludins protein-1 (ZO-1). Therefore, dietary supplementation of EB may increase intestinal barrier function, tight junction protein expression, anti-inflammatory cytokines, and decrease proinflammatory cytokines synthesis in IPEC-J2 cells.
Collapse
Affiliation(s)
- Dongsheng Che
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Bao Zhao
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yueli Fan
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Rui Han
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chun Zhang
- College of Animal Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Guixin Qin
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Seidu Adams
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Hailong Jiang
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
6
|
Wang C, Li Y, Chen B, Zou M. In vivo pharmacokinetics, biodistribution and the anti-tumor effect of cyclic RGD-modified doxorubicin-loaded polymers in tumor-bearing mice. Colloids Surf B Biointerfaces 2016; 146:31-8. [PMID: 27244048 DOI: 10.1016/j.colsurfb.2016.05.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/14/2016] [Accepted: 05/17/2016] [Indexed: 11/17/2022]
Abstract
In our previous study, we successfully produced and characterized a multifunctional drug delivery system with doxorubicin (RC/GO/DOX), which was based on graphene oxide (GO) and cyclic RGD-modified chitosan (RC). Its characteristics include: pH-responsiveness, active targeting of hepatocarcinoma cells, and efficient loading with controlled drug release. Here, we report the pharmacokinetics, biodistribution, and anti-tumor efficacy of RC/GO/DOX polymers in tumor-bearing nude mice. The objective of this study is to assess its targeting potential for tumors. Pharmacokinetic and biodistribution profiles demonstrated that tumor accumulation of RC/GO/DOX polymers was almost three times higher than the others, highlighting the efficacy of the active targeting strategy. Furthermore, the tumor inhibition rate of RC/GO/DOX polymers was 56.64%, 2.09 and 2.93 times higher than that of CS/GO/DOX polymers (without modification) and the DOX solution, respectively. Anti-tumor efficacy results indicated that the tumor growth was better controlled by RC/GO/DOX polymers than the others. Hematoxylin and eosin (H&E) staining showed remarkable changes in tumor histology. Compared with the saline group, the tumor section from the RC/GO/DOX group revealed a marked increase in the quantity of apoptotic and necrotic cells, and a reduction in the quantity of the blood vessels. Together, these studies show that this new system could be regarded as a suitable form of DOX-based treatment of the hepatocellular carcinoma.
Collapse
Affiliation(s)
- Chen Wang
- School of Pharmacy, Xiamen Medical College, Xiamen 361008, PR China.
| | - Yuan Li
- School of Pharmacy, Xiamen Medical College, Xiamen 361008, PR China
| | - Binbin Chen
- Department of Pharmacy, Xiamen Xianyue Hospital, 361012, PR China
| | - Meijuan Zou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
7
|
Yellepeddi VK, Ghandehari H. Poly(amido amine) dendrimers in oral delivery. Tissue Barriers 2016; 4:e1173773. [PMID: 27358755 DOI: 10.1080/21688370.2016.1173773] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 01/11/2023] Open
Abstract
Poly(amidoamine) (PAMAM) dendrimers have been extensively investigated for oral delivery applications due to their ability to translocate across the gastrointestinal epithelium. In this Review, we highlight recent advances in the evaluation of PAMAM dendrimers as oral drug delivery carriers. Specifically, toxicity, mechanisms of transepithelial transport, models of the intestinal epithelial barrier including isolated human intestinal tissue model, detection of dendrimers, and surface modification are discussed. We also highlight evaluation of various PAMAM dendrimer-drug conjugates for their ability to transport across gastrointestinal epithelium for improved oral bioavailability. In addition, current challenges and future trends for clinical translation of PAMAM dendrimers as carriers for oral delivery are discussed.
Collapse
Affiliation(s)
- Venkata K Yellepeddi
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
8
|
Li J, Chen L, Liu N, Li S, Hao Y, Zhang X. EGF-coated nano-dendriplexes for tumor-targeted nucleic acid delivery in vivo. Drug Deliv 2015; 23:1718-25. [PMID: 25693638 DOI: 10.3109/10717544.2015.1004381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The clinical success of therapeutic DNA is still hindered due to the lack of effective delivery carriers. Here, we designed a tumor-targeted gene nano delivery system based on EGFR targeting strategy. Epidermal growth factor (EGF) was introduced to nano-complexes of PAMAM dendrimer and DNA via electrostatic interactions to form self-assembled PAMAM/DNA/EGF nano-complexes. The properties of self-assembled complexes were characterized by gel retardation assay and particle size and zeta potential analysis. Meanwhile, the toxicity of EGF-dendriplexes was evaluated by the MTT assay, which indicated that the complexes exhibited decreased cytotoxicity with the incorporation of EGF. We labeled polyamidoamine (PAMAM) dendrimers with FITC or a near-infrared (NIR) dye Lss670 and tested the cellular uptake in vitro and biodistribution in xenograft mouse tumor models. As compared to dendriplexes, the ternary EGF-dendriplexes showed a significantly higher cellular uptake into HepG2 cells due to the specific binding between EGF and EGF receptor (EGFR) over expressed on HepG2 cells, which resulted in the enhanced gene transfection efficiency. The biodistribution of EGF-dendriplexes in vivo was monitored with in vivo imaging technique, which indicated that EGF-dendriplexes enhanced EGFR-positive tumor-targeted biodistribution. These findings indicate that this novel nano-vector realized efficiently tumor-targeting gene delivery and high efficient gene expression in vivo, and it may possess a potential targeting gene delivery system in cancer therapy.
Collapse
Affiliation(s)
- Jun Li
- a School of Medicine, Tsinghua University , Beijing , China
| | - Lei Chen
- b Department of Gynaecology and Obstetrics , PLA Navy General Hospital , Beijing , China , and
| | - Nan Liu
- a School of Medicine, Tsinghua University , Beijing , China
| | - Shengnan Li
- b Department of Gynaecology and Obstetrics , PLA Navy General Hospital , Beijing , China , and
| | - Yanli Hao
- a School of Medicine, Tsinghua University , Beijing , China
| | - Xiaoning Zhang
- a School of Medicine, Tsinghua University , Beijing , China .,c Collaborative Innovation Center for Biotherapy, Tsinghua University , Beijing , China
| |
Collapse
|
9
|
Bharatwaj B, Mohammad AK, Dimovski R, Cassio FL, Bazito RC, Conti D, Fu Q, Reineke J, da Rocha SRP. Dendrimer nanocarriers for transport modulation across models of the pulmonary epithelium. Mol Pharm 2015; 12:826-38. [PMID: 25455560 PMCID: PMC4350608 DOI: 10.1021/mp500662z] [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] [Indexed: 11/29/2022]
Abstract
![]()
The purpose of this study was to
determine the effect of PEGylation
on the interaction of poly(amidoamine) (PAMAM) dendrimer nanocarriers
(DNCs) with in vitro and in vivo models of the pulmonary epithelium. Generation-3 PAMAM dendrimers
with varying surface densities of PEG 1000 Da were synthesized and
characterized. The results revealed that the apical to basolateral
transport of DNCs across polarized Calu-3 monolayers increases with
an increase in PEG surface density. DNC having the greatest number
of PEG groups (n = 25) on their surface traversed
at a rate 10-fold greater than its non-PEGylated counterpart, in spite
of their larger size. This behavior was attributed to a significant
reduction in charge density upon PEGylation. We also observed that
PEGylation can be used to modulate cellular internalization. The total
uptake of PEG-free DNC into polarized Calu-3 monolayers was 12% (w/w)
vs 2% (w/w) for that with 25 PEGs. Polarization is also shown to be
of great relevance in studying this in vitro model
of the lung epithelium. The rate of absorption of DNCs administered
to mice lungs increased dramatically when conjugated with 25 PEG groups,
thus supporting the in vitro results. The exposure
obtained for the DNC with 25PEG was determined to be very high, with
peak plasma concentrations reaching 5 μg·mL–1 within 3 h. The combined in vitro and in
vivo results shown here demonstrate that PEGylation can be
potentially used to modulate the internalization and transport of
DNCs across the pulmonary epithelium. Modified dendrimers thereby
may serve as a valuable platform that can be tailored to target the
lung tissue for treating local diseases, or the circulation, using
the lung as pathway to the bloodstream, for systemic delivery.
Collapse
Affiliation(s)
- Balaji Bharatwaj
- Department of Chemical Engineering and Materials Science, and ‡Department of Pharmaceutical Sciences, Wayne State University , Detroit, Michigan 48202, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Bergin IL, Witzmann FA. Nanoparticle toxicity by the gastrointestinal route: evidence and knowledge gaps. INTERNATIONAL JOURNAL OF BIOMEDICAL NANOSCIENCE AND NANOTECHNOLOGY 2013; 3:10.1504/IJBNN.2013.054515. [PMID: 24228068 PMCID: PMC3822607 DOI: 10.1504/ijbnn.2013.054515] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The increasing interest in nanoparticles for advanced technologies, consumer products, and biomedical applications has led to great excitement about potential benefits but also concern over the potential for adverse human health effects. The gastrointestinal tract represents a likely route of entry for many nanomaterials, both directly through intentional ingestion or indirectly via nanoparticle dissolution from food containers or by secondary ingestion of inhaled particles. Additionally, increased utilisation of nanoparticles may lead to increased environmental contamination and unintentional ingestion via water, food animals, or fish. The gastrointestinal tract is a site of complex, symbiotic interactions between host cells and the resident microbiome. Accordingly, evaluation of nanoparticles must take into consideration not only absorption and extraintestinal organ accumulation but also the potential for altered gut microbes and the effects of this perturbation on the host. The existing literature was evaluated for evidence of toxicity based on these considerations. Focus was placed on three categories of nanomaterials: nanometals and metal oxides, carbon-based nanoparticles, and polymer/dendrimers with emphasis on those particles of greatest relevance to gastrointestinal exposures.
Collapse
Affiliation(s)
- Ingrid L. Bergin
- Unit for Laboratory Animal Medicine, University of Michigan, 1150 W. Medical Center Dr, 018 ARF, Ann Arbor, MI 48197, USA,
| | - Frank A. Witzmann
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 1345 West 16th Street, Indianapolis IN 46202, USA
| |
Collapse
|
11
|
Lodemann U, Einspanier R, Scharfen F, Martens H, Bondzio A. Effects of zinc on epithelial barrier properties and viability in a human and a porcine intestinal cell culture model. Toxicol In Vitro 2012; 27:834-43. [PMID: 23274768 DOI: 10.1016/j.tiv.2012.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 12/14/2012] [Accepted: 12/19/2012] [Indexed: 01/01/2023]
Abstract
Zinc is an essential trace element with a variety of physiological and biochemical functions. Piglets are commonly supplemented, during the weaning period, with doses of zinc above dietary requirements with positive effects on health and performance that might be attributed to anti-secretory and barrier-enhancing effects in the intestine. For a better understanding of these observations increasing zinc sulfate (ZnSO4; 0-200μM) concentrations were used in an in vitro culture model of porcine (IPEC-J2) and human (Caco-2) intestinal epithelial cells and effects on barrier function, viability, and the mRNA expression of one selected heat shock protein (Hsp) were assessed. When treated apically with zinc sulfate, the transepithelial electrical resistance (TEER) did not change significantly. In contrast, cell viability measured by lactate dehydrogenase (LDH) leakage, by ATP and by WST-1 conversion in postconfluent IPEC-J2 monolayers was affected after a 24-h treatment with 200μM ZnSO4. Caco-2 cells were more resistant to Zn. ZnSO4 did not induce any effect on viability, except when it was used at the highest concentration (200μM), and only in preconfluent cells. Furthermore, ZnSO4 induced Hsp70 mRNA expression at 200μM and was more pronounced in preconfluent cells. The observed dose-related effects of zinc are cell-line specific and depended on the differentiation status of the cells. The IPEC-J2 cell line appears to be a suitable in vitro model to characterize specific effects on porcine intestinal cells.
Collapse
Affiliation(s)
- U Lodemann
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany.
| | | | | | | | | |
Collapse
|
12
|
Chapman JC, Liu Y, Zhu L, Rhoads JM. Arginine and citrulline protect intestinal cell monolayer tight junctions from hypoxia-induced injury in piglets. Pediatr Res 2012; 72:576-82. [PMID: 23041662 PMCID: PMC3976428 DOI: 10.1038/pr.2012.137] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Arginine (Arg) is deficient in the serum of the preterm neonate and is lower in those developing intestinal ischemia. We investigated whether Arg or its precursor, citrulline (Cit), protects intestinal tight junctions (TJs) from hypoxia (HX) and determined whether inducible nitric oxide (NO) plays a role. METHODS Neonatal piglet jejunal IPEC-J2 cell monolayers were treated with Arg or Cit, reversible and irreversible NO synthetase (NOS) inhibitors, and were exposed to HX. TJs were assessed by serial measurements of transepithelial electrical resistance (TEER), flux of inulin-fluorescein isothiocyanate, and immunofluorescent staining of TJ proteins. RESULTS We found that Arg and Cit were protective against HX-related damage. At the final time point (14 h), the mean TEER ratio (TEER as compared with baseline) for Arg + HX and Cit + HX was significantly higher than that for HX alone. Both Arg and Cit were associated with decreased inulin flux across hypoxic monolayers and qualitatively preserved TJ proteins. Irreversible inhibition of NOS blocked this protective effect. Lipid peroxidation assay showed that our model did not produce oxidant injury. CONCLUSION Arg and Cit, via a mechanism dependent on NO donation, protected intestinal epithelial integrity.
Collapse
Affiliation(s)
- John C. Chapman
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center, Houston, Texas
| | - Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, Pediatric Research Institute, University of Texas Health Science Center, Houston, Texas
| | - Limin Zhu
- Department of Pediatrics, Division of Gastroenterology, Pediatric Research Institute, University of Texas Health Science Center, Houston, Texas
| | - J. Marc Rhoads
- Department of Pediatrics, Division of Gastroenterology, Pediatric Research Institute, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
13
|
Kaminskas LM, Boyd BJ, Porter CJH. Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties. Nanomedicine (Lond) 2012; 6:1063-84. [PMID: 21955077 DOI: 10.2217/nnm.11.67] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dendrimers show increasing promise as drug-delivery vectors and can be generated with a wide range of scaffold structures, sizes and surface functionalities. To this point, the majority of studies of dendrimer-based drug-delivery systems have detailed pharmacodynamic outcomes, or have followed the pharmacokinetics of a solubilized or conjugated drug. By contrast, detailed commentary on the in vivo fate of the dendrimer carrier is less evident, even though the pharmacokinetics of the carrier will likely dictate both pharmacodynamic and toxicokinetic outcomes. In the current article, the influence of size, structure and surface functionality on the absorption, distribution, metabolism and elimination (ADME) properties of dendrimers have been examined and the implications of these findings for delivery system design are discussed.
Collapse
Affiliation(s)
- Lisa M Kaminskas
- Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University. 381 Royal Parade, Parkville, VIC, 3052, Australia
| | | | | |
Collapse
|
14
|
Sadekar S, Ghandehari H. Transepithelial transport and toxicity of PAMAM dendrimers: implications for oral drug delivery. Adv Drug Deliv Rev 2012; 64:571-88. [PMID: 21983078 DOI: 10.1016/j.addr.2011.09.010] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 08/29/2011] [Accepted: 09/02/2011] [Indexed: 12/17/2022]
Abstract
This article summarizes efforts to evaluate poly(amido amine) (PAMAM) dendrimers as carriers for oral drug delivery. Specifically, the effect of PAMAM generation, surface charge and surface modification on toxicity, cellular uptake and transepithelial transport is discussed. Studies on Caco-2 monolayers, as models of intestinal epithelial barrier, show that by engineering surface chemistry of PAMAM dendrimers, it is possible to minimize toxicity while maximizing transepithelial transport. It has been demonstrated that PAMAM dendrimers are transported by a combination of paracellular and transcellular routes. Depending on surface chemistry, PAMAM dendrimers can open the tight junctions of epithelial barriers. This tight junction opening is in part mediated by internalization of the dendrimers. Transcellular transport of PAMAM dendrimers is mediated by a variety of endocytic mechanisms. Attachment or complexation of cytotoxic agents to PAMAM dendrimers enhances the transport of such drugs across epithelial barriers. A remaining challenge is the design and development of linker chemistries that are stable in the gastrointestinal tract (GIT) and the blood stream, but amenable to cleavage at the target site of action. Recent efforts have focused on the use of PAMAM dendrimers as penetration enhancers. Detailed in vivo oral bioavailability of PAMAM dendrimer-drug conjugates, as a function of physicochemical properties will further need to be assessed.
Collapse
|
15
|
Card JW, Jonaitis TS, Tafazoli S, Magnuson BA. An appraisal of the published literature on the safety and toxicity of food-related nanomaterials. Crit Rev Toxicol 2010; 41:22-49. [DOI: 10.3109/10408444.2010.524636] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
16
|
Yellepeddi VK, Kumar A, Palakurthi S. Surface modified poly(amido)amine dendrimers as diverse nanomolecules for biomedical applications. Expert Opin Drug Deliv 2009; 6:835-50. [DOI: 10.1517/17425240903061251] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|