451
|
Song H, Canup BSB, Ngo VL, Denning TL, Garg P, Laroui H. Internalization of Garlic-Derived Nanovesicles on Liver Cells is Triggered by Interaction With CD98. ACS OMEGA 2020; 5:23118-23128. [PMID: 32954162 PMCID: PMC7495725 DOI: 10.1021/acsomega.0c02893] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/11/2020] [Indexed: 05/20/2023]
Abstract
The mechanism of how plant-derived nanovesicles are uptaken by cells remains unknown. In this study, the garlic-derived nanovesicles (GDVs) were isolated and digested with trypsin to remove all surface proteins. Digested GDVs showed less uptake compared to undigested GDVs, confirming that the surface proteins played a role in the endocytosis. On the cell side (HepG2), interestingly, blocking the CD98 receptors significantly reduced the uptake of GDVs. During the cellular internalization of GDVs, we observed that some surface proteins of GDVs were co-localized with CD98. A total lysate of the GDV surface showed a high presence of a mannose-specific binding protein, II lectin. Blocking GDV II lectin (using mannose preincubation) highly reduced the GDV internalization, which supports that direct interaction between II lectin and CD98 plays an important role in internalization. The GDVs also exhibited in vitro anti-inflammatory effect by downregulating proinflammatory factors on the HepG2 cells. This work contributes to understanding a part of the GDV internalization process and the cellular anti-inflammatory effects of garlic.
Collapse
Affiliation(s)
- Heliang Song
- Department
of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, Georgia 30302, United States
| | - Brandon S. B. Canup
- Department
of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, Georgia 30302, United States
| | - Vu L. Ngo
- Department
of Biology, Institute for Biomedical Sciences (IBMS), Georgia State University, Atlanta, Georgia 30302, United States
| | - Timothy L. Denning
- Department
of Biology, Institute for Biomedical Sciences (IBMS), Georgia State University, Atlanta, Georgia 30302, United States
| | - Pallavi Garg
- Department
of Biology, Institute for Biomedical Sciences (IBMS), Georgia State University, Atlanta, Georgia 30302, United States
| | - Hamed Laroui
- Department
of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, Georgia 30302, United States
| |
Collapse
|
452
|
Bi K, Zhang X, Chen W, Diao H. MicroRNAs Regulate Intestinal Immunity and Gut Microbiota for Gastrointestinal Health: A Comprehensive Review. Genes (Basel) 2020; 11:genes11091075. [PMID: 32932716 PMCID: PMC7564790 DOI: 10.3390/genes11091075] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are small non-coding RNAs regulating gene expression at the post-transcriptional level. The regulation of microRNA expression in the gut intestine is gradually recognized as one of the crucial contributors of intestinal homeostasis and overall health. Recent studies indicated that both the microRNAs endogenous in the gut intestine and exogenous from diets could play influential roles in modulating microbial colonization and intestinal immunity. In this review, we discuss the biological functions of microRNAs in regulating intestinal homeostasis by modulating intestinal immune responses and gut microbiota. We particularly focus on addressing the microRNA-dependent communication and interactions among microRNA, gut microbiota, and intestinal immune system. Besides, we also summarize the roles of diet-derived microRNAs in host-microbiome homeostasis and their benefits on intestinal health. A better understanding of the relationships among intestinal disorders, microRNAs, and other factors influencing intestinal health can facilitate the application of microRNA-based therapeutics for gastrointestinal diseases.
Collapse
|
453
|
Berger E, Colosetti P, Jalabert A, Meugnier E, Wiklander OP, Jouhet J, Errazurig-Cerda E, Chanon S, Gupta D, Rautureau GJ, Geloen A, El-Andaloussi S, Panthu B, Rieusset J, Rome S. Use of Nanovesicles from Orange Juice to Reverse Diet-Induced Gut Modifications in Diet-Induced Obese Mice. Mol Ther Methods Clin Dev 2020; 18:880-892. [PMID: 32953937 PMCID: PMC7481887 DOI: 10.1016/j.omtm.2020.08.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023]
Abstract
We have determined whether orange juice-derived nanovesicles (ONVs) could be used for the treatment of obesity-associated intestinal complications. ONVs were characterized by lipidomic, metabolomic, electron microscopy. In vitro, intestinal barriers (IBs = Caco-2+HT-29-MTX) were treated with ONVs and co-cultured with adipocytes to monitor IB fat release. In vivo, obesity was induced with a high-fat, high-sucrose diet (HFHSD mice) for 12 weeks. Then, half of HFHSD mice were gavaged with ONVs. One-month ONV treatment did not modify HFHSD-induced insulin resistance but reversed diet-induced gut modifications. In the jejunum, ONVs increased villi size, reduced triglyceride content, and modulated mRNA levels of genes involved in immune response (tumor necrosis factor [TNF]-α and interleukin [IL]-1β), barrier permeability (CLDN1, OCLN, ZO1), fat absorption, and chylomicron release. ONVs targeted microsomal triglyceride transfer protein (MTP) and angiopoietin-like protein-4 (ANGPTL4), two therapeutic targets to reduce plasma lipids and inflammation in gastrointestinal diseases. Interestingly, ONV treatment did not aggravate liver steatosis, as MTP mRNA was increased in the liver. Therefore, ONVs protected both intestine and the liver from fat overload associated with the HFHSD. As ONVs concentrated amino acids and bioactive lipids versus orange juice, which are deficient in obese patients, the use of ONVs as a dietary supplement could bring physiological relevant compounds in the jejunum to accelerate the restoration of intestinal functions during weight loss in obese patients.
Collapse
Affiliation(s)
- Emmanuelle Berger
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Pascal Colosetti
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Audrey Jalabert
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Emmanuelle Meugnier
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Oscar P.B. Wiklander
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire & Végétale (LPCV), CNRS (UMR5168)/Université Grenoble Alpes/INRAe (UMR1417)/CEA Grenoble, Institut de Biosciences et Biotechnologies de Grenoble, France
| | | | - Stéphanie Chanon
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Dhanu Gupta
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Gilles J.P. Rautureau
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, ENS de Lyon, Centre de RMN à Très Hauts Champs (CRMN), FRE 2034, Villeurbanne, France
| | - Alain Geloen
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Samir El-Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Baptiste Panthu
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Jennifer Rieusset
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| | - Sophie Rome
- CarMeN Laboratory (INRAe U1397, INSERM U1060, Lyon 1 University, INSA Lyon), Bâtiment CENS ELI-2D, Pierre-Bénite, France
| |
Collapse
|
454
|
New Insights of Oral Colonic Drug Delivery Systems for Inflammatory Bowel Disease Therapy. Int J Mol Sci 2020; 21:ijms21186502. [PMID: 32899548 PMCID: PMC7555849 DOI: 10.3390/ijms21186502] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Colonic Drug Delivery Systems (CDDS) are especially advantageous for local treatment of inflammatory bowel diseases (IBD). Site-targeted drug release allows to obtain a high drug concentration in injured tissues and less systemic adverse effects, as consequence of less/null drug absorption in small intestine. This review focused on the reported contributions in the last four years to improve the effectiveness of treatments of inflammatory bowel diseases. The work concludes that there has been an increase in the development of CDDS in which pH, specific enzymes, reactive oxygen species (ROS), or a combination of all of these triggers the release. These delivery systems demonstrated a therapeutic improvement with fewer adverse effects. Future perspectives to the treatment of this disease include the elucidation of molecular basis of IBD diseases in order to design more specific treatments, and the performance of more in vivo assays to validate the specificity and stability of the obtained systems.
Collapse
|
455
|
Feng K, Wei YS, Hu TG, Linhardt RJ, Zong MH, Wu H. Colon-targeted delivery systems for nutraceuticals: A review of current vehicles, evaluation methods and future prospects. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2020.05.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
456
|
Liu Y, Wu S, Koo Y, Yang A, Dai Y, Khant H, Osman SR, Chowdhury M, Wei H, Li Y, Court K, Hwang E, Wen Y, Dasari SK, Nguyen M, Tang ECC, Chehab EW, de Val N, Braam J, Sood AK. Characterization of and isolation methods for plant leaf nanovesicles and small extracellular vesicles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102271. [PMID: 32702466 DOI: 10.1016/j.nano.2020.102271] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 05/27/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022]
Abstract
Mammalian small extracellular vesicles (sEVs) can deliver diverse molecules to target cells. However, they are difficult to obtain in large quantities and can activate host immune responses. Plant-derived vesicles may help to overcome these challenges. We optimized isolation methods for two types of plant vesicles, nanovesicles from disrupted leaf and sEVs from the extracellular apoplastic space of Arabidopsis thaliana. Both preparations yielded intact vesicles of uniform size, and a mean membrane charge of approximately -25 mV. We also demonstrated applicability of these preparative methods using Brassicaceae vegetables. Proteomic analysis of a subset of vesicles with a density of 1.1-1.19 g mL-1 sheds light on the likely cellular origin and complexity of the vesicles. Both leaf nanovesicles and sEVs were taken up by cancer cells, with sEVs showing an approximately three-fold higher efficiency compared to leaf nanovesicles. These results support the potential of plant-derived vesicles as vehicles for therapeutic delivery.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; BioSciences, Rice University, Houston, TX.
| | - Sherry Wu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | | | - An Yang
- BioSciences, Rice University, Houston, TX; State Key Laboratory of Vegetation and Environmental Change, Institute of Botany, Chinese Academy of Sciences, Beijing, People's Republic of China.
| | - Yanwan Dai
- BioSciences, Rice University, Houston, TX.
| | - Htet Khant
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, Frederick, MD; Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Inc., Frederick, MD.
| | | | - Mamur Chowdhury
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX.
| | - Yang Li
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Karem Court
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | | | - Yunfei Wen
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Santosh K Dasari
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | | | | | | | - Natalia de Val
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, Frederick, MD; Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Inc., Frederick, MD.
| | | | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
457
|
Lipid-Based Drug Delivery Nanoplatforms for Colorectal Cancer Therapy. NANOMATERIALS 2020; 10:nano10071424. [PMID: 32708193 PMCID: PMC7408503 DOI: 10.3390/nano10071424] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is a prevalent disease worldwide, and patients at late stages of CRC often suffer from a high mortality rate after surgery. Adjuvant chemotherapeutics (ACs) have been extensively developed to improve the survival rate of such patients, but conventionally formulated ACs inevitably distribute toxic chemotherapeutic drugs to healthy organs and thus often trigger severe side effects. CRC cells may also develop drug resistance following repeat dosing of conventional ACs, limiting their effectiveness. Given these limitations, researchers have sought to use targeted drug delivery systems (DDSs), specifically the nanotechnology-based DDSs, to deliver the ACs. As lipid-based nanoplatforms have shown the potential to improve the efficacy and safety of various cytotoxic drugs (such as paclitaxel and vincristine) in the clinical treatment of gastric cancer and leukemia, the preclinical progress of lipid-based nanoplatforms has attracted increasing interest. The lipid-based nanoplatforms might be the most promising DDSs to succeed in entering a clinical trial for CRC treatment. This review will briefly examine the history of preclinical research on lipid-based nanoplatforms, summarize the current progress, and discuss the challenges and prospects of using such approaches in the treatment of CRC.
Collapse
|
458
|
Yang C, Zhang M, Sung J, Wang L, Jung Y, Merlin D. Autologous Exosome Transfer: A New Personalised Treatment Concept to Prevent Colitis in a Murine Model. J Crohns Colitis 2020; 14:841-855. [PMID: 31710674 PMCID: PMC7346889 DOI: 10.1093/ecco-jcc/jjz184] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Epigenetic information delivered by intestinal exosomes can be useful for diagnosing intestinal diseases, such as ulcerative colitis, but the therapeutic effects of intestinal exosomes have not been fully exploited. We herein developed an autologous exosome therapy that could treat intestinal disease without any risk of inducing a systemic immunological reaction. METHODS Intestinal exosomes were isolated and purified from faeces by our newly developed multi-step sucrose gradient ultracentrifugation method. Lipopolysaccharide [LPS]-activated macrophages were employed to test the in vitro anti-inflammatory ability of intestinal exosomes. To evaluate the in vivo anti-inflammatory activity of our system, we gavaged dextran sulphate sodium [DSS]-induced colitic mice with their own healing phase intestinal exosomes. RESULTS Mouse intestinal exosomes are round extracellular vesicles with a hydrodynamic diameter of ~140 [±20] nm and a surface charge of ~-12 [±3] mV. Among the exosomes obtained at four different stages of DSS-induced ulcerative colitis [1, before treatment; 2, DSS-treated; 3, healing phase; and 4, back to normal], the healing phase exosomes showed the best in vitro anti-inflammatory effects and promotion of wound healing. Moreover, oral co-administration of autologous healing phase exosomes with DSS was found to significantly reduce the risk of a second round of DSS-induced ulcerative colitis in mice. CONCLUSIONS Intestinal exosomes obtained during the healing phase that follows induced intestinal inflammation could strongly promote wound healing in the host. Oral administration of autologous exosomes from the healing phase could be a safe and effective approach for treating the ulcerative colitis of a given patient in the context of personalised medicine.
Collapse
Affiliation(s)
- Chunhua Yang
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Mingzhen Zhang
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Junsik Sung
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Lixin Wang
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
- Atlanta Veterans Medical Center, Decatur, GA, USA
| | - Yunjin Jung
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Didier Merlin
- Digestive Disease Research Group, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
- Atlanta Veterans Medical Center, Decatur, GA, USA
| |
Collapse
|
459
|
Zhu Y, Zhang G, Li M, Ma L, Huang J, Qiu L. Ultrasound-Augmented Phase Transition Nanobubbles for Targeted Treatment of Paclitaxel-Resistant Cancer. Bioconjug Chem 2020; 31:2008-2020. [PMID: 32628454 DOI: 10.1021/acs.bioconjchem.0c00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yi Zhu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Ultrasound, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Guonan Zhang
- Department of Gynecological Oncology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Meiying Li
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Lang Ma
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Huang
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
460
|
Shukurova MK, Asikin Y, Chen Y, Kusano M, Watanabe KN. Profiling of Volatile Organic Compounds in Wild Indigenous Medicinal Ginger ( Zingiber barbatum Wall.) from Myanmar. Metabolites 2020; 10:E248. [PMID: 32549365 PMCID: PMC7344531 DOI: 10.3390/metabo10060248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 11/17/2022] Open
Abstract
The emissions of volatile organic compounds (VOCs) strongly depend on the plant species and are differently represented in specific taxa. VOCs have a degree of chemical diversity and also can serve as chemotaxonomic markers. Zingiber barbatum Wall. is a wild medicinal ginger plant endemic to Myanmar whose VOC composition has never been screened before. In this study, we screened the rhizome of Z. barbatum to identify the VOC composition by the application of gas chromatography combined with time-of-flight-mass spectrometry (GC-TOF-MS). The resulting VOC profile of Z. barbatum showed that it consists mainly of monoterpenes (21%) and sesquiterpenes (30%). Intraspecific similarities and dissimilarities were found to exist between Z. barbatum genotypes in terms of VOC composition. Four accessions (ZO191, ZO223, ZO217, and the control accession ZO105) collected from the Shan State and Mandalay region of Myanmar were found to share a similar VOC profile, while two accessions (ZO64 and ZO160) collected from the Bago region were found to vary in their VOC profiles compared with the control accession. The two identified compounds, i.e., α-bergamotene and β-(E)-guaiene may serve as discriminative chemical markers for the characterization of Z. barbatum species collected in these three geographical regions of Myanmar. This study represents a first attempt to identify and describe the VOCs in the medicinal species Z. barbatum that have not been reported to date.
Collapse
Affiliation(s)
- Musavvara Kh. Shukurova
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan;
| | - Yonathan Asikin
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Okinawa 903-0213, Japan;
| | - Yanhang Chen
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan;
| | - Miyako Kusano
- Faculty of Life and Environmental Science, University of Tsukuba, Ibaraki 305-8572, Japan; (M.K.); (K.N.W.)
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, Ibaraki 305-8572, Japan
- RIKEN Center for Sustainable Resource Science, Kanagawa 230-0045, Japan
| | - Kazuo N. Watanabe
- Faculty of Life and Environmental Science, University of Tsukuba, Ibaraki 305-8572, Japan; (M.K.); (K.N.W.)
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, Ibaraki 305-8572, Japan
| |
Collapse
|
461
|
Khare T, Palakurthi SS, Shah BM, Palakurthi S, Khare S. Natural Product-Based Nanomedicine in Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:E3956. [PMID: 32486445 PMCID: PMC7312938 DOI: 10.3390/ijms21113956] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
: Many synthetic drugs and monoclonal antibodies are currently in use to treat Inflammatory Bowel Disease (IBD). However, they all are implicated in causing severe side effects and long-term use results in many complications. Numerous in vitro and in vivo experiments demonstrate that phytochemicals and natural macromolecules from plants and animals reduce IBD-related complications with encouraging results. Additionally, many of them modify enzymatic activity, alleviate oxidative stress, and downregulate pro-inflammatory transcriptional factors and cytokine secretion. Translational significance of natural nanomedicine and strategies to investigate future natural product-based nanomedicine is discussed. Our focus in this review is to summarize the use of phytochemicals and macromolecules encapsulated in nanoparticles for the treatment of IBD and IBD-associated colorectal cancer.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Brijesh M. Shah
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
| |
Collapse
|
462
|
Tian R, Liu X, Luo Y, Jiang S, Liu H, You F, Zheng C, Wu J. Apoptosis Exerts a Vital Role in the Treatment of Colitis-Associated Cancer by Herbal Medicine. Front Pharmacol 2020; 11:438. [PMID: 32410986 PMCID: PMC7199713 DOI: 10.3389/fphar.2020.00438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/20/2020] [Indexed: 12/24/2022] Open
Abstract
Colitis-associated cancer (CAC) is known as inflammatory bowel disease (IBD)-developed colorectal cancer, the pathogenesis of which involves the occurrence of apoptosis. Western drugs clinically applied to CAC are often single-targeted and exert many adverse reactions after long-term administration, so it is urgent to develop new drugs for the treatment of CAC. Herbal medicines commonly have multiple components with multiple targets, and most of them are low-toxicity. Some herbal medicines have been reported to ameliorate CAC through inducing apoptosis, but there is still a lack of systematic review. In this work, we reviewed articles published in Sci Finder, Web of Science, PubMed, Google Scholar, CNKI, and other databases in recent years by setting the keywords as apoptosis in combination with colitis-associated cancer. We summarized the herbal medicine extracts or their compounds that can prevent CAC by modulating apoptosis and analyzed the mechanism of action. The results show the following. (1) Herbal medicines regulate both the mitochondrial apoptosis pathway and death receptor apoptosis pathway. (2) Herbal medicines modulate the above two apoptotic pathways by affecting signal transductions of IL-6/STAT3, MAPK/NF-κ B, Oxidative stress, Non-canonical TGF-β1, WNT/β-catenin, and Cell cycle, thereby ameliorating CAC. We conclude that following. (1) Studies on the role of herbal medicine in regulating apoptosis through the Ras/Raf/ERK, WNT/β-catenin, and Cell cycle pathways have not yet been carried out in sufficient depth. (2) The active constituents of reported anti-CAC herbal medicine mainly include polyphenols, terpenoids, and saccharide. Also, we identified other herbal medicines with the constituents mentioned above as their main components, aiming to provide a reference for the clinical use of herbal medicine in the treatment of CAC. (3) New dosage forms can be utilized to elevate the targeting and reduce the toxicity of herbal medicine.
Collapse
Affiliation(s)
- Ruimin Tian
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacology, North Sichuan Medical College, Nanchong, China
| | - Xianfeng Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanqin Luo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengnan Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiasi Wu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
463
|
Nouri Z, Hajialyani M, Izadi Z, Bahramsoltani R, Farzaei MH, Abdollahi M. Nanophytomedicines for the Prevention of Metabolic Syndrome: A Pharmacological and Biopharmaceutical Review. Front Bioeng Biotechnol 2020; 8:425. [PMID: 32478050 PMCID: PMC7240035 DOI: 10.3389/fbioe.2020.00425] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome includes a series of metabolic abnormalities that leads to diabetes mellitus and cardiovascular diseases. Plant extracts, due to their unique advantages like anti-inflammatory, antioxidant, and insulin sensitizing properties, are interesting therapeutic options to manage MetS; however, the poor solubility and low bioavailability of lipophilic bioactive components in the herbal extracts are two critical challenges. Nano-scale delivery systems are suitable to improve delivery of herbal extracts. This review, for the first time, focuses on nanoformulations of herbal extracts in MetS and related complications. Included studies showed that several forms of nano drug delivery systems such as nanoemulsions, solid lipid nanoparticles, nanobiocomposites, and green-synthesized silver, gold, and zinc oxide nanoparticles have been developed using herbal extracts. It was shown that the method of preparation and related parameters such as temperature and type of polymer are important factors affecting physicochemical stability and therapeutic activity of the final product. Many of these formulations could successfully decrease the lipid profile, inflammation, oxidative damage, and insulin resistance in in vitro and in vivo models of MetS-related complications. Further studies are still needed to confirm the safety and efficacy of these novel herbal formulations for clinical application.
Collapse
Affiliation(s)
- Zeinab Nouri
- Students Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marziyeh Hajialyani
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roodabeh Bahramsoltani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- PhytoPharmacology Interest Group, Universal Scientific Education and Research Network, Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
464
|
Munir J, Lee M, Ryu S. Exosomes in Food: Health Benefits and Clinical Relevance in Diseases. Adv Nutr 2020; 11:687-696. [PMID: 31796948 PMCID: PMC7231590 DOI: 10.1093/advances/nmz123] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/14/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Exosomes are membrane-bound organelles generally secreted by eukaryotic cells that contain mRNAs, microRNAs, and/or proteins. However, recent studies have reported the isolation of these particles from foods such as lemon, ginger, and milk. Owing to their absorption by intestinal cells and further travel via the bloodstream, exosomes can reach distant organs and affect overall health in both infants and adults. The potential role of food-derived exosomes (FDEs) in alleviating diseases, as well as in modulating the gut microbiota has been shown, but the underlying mechanism is still unknown. Moreover, exosomes may provide biocompatible vehicles for the delivery of anti-cancer drugs, such as doxorubicin. Thus, exosomes may allow medical nutritionists and clinicians to develop safe and targeted therapies for the treatment of various pathologies. The present review introduces FDEs and their contents, highlights their role in disease and infant/adult health, and explores their potential use as therapeutic agents.
Collapse
Affiliation(s)
| | - Mihye Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Chungcheongnam-do, South Korea
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-bioscience (SIMS), Soonchunhyang University, Cheonan, Chungcheongnam-do, South Korea
| |
Collapse
|
465
|
Aggarwal V, Tuli HS, Kaur J, Aggarwal D, Parashar G, Chaturvedi Parashar N, Kulkarni S, Kaur G, Sak K, Kumar M, Ahn KS. Garcinol Exhibits Anti-Neoplastic Effects by Targeting Diverse Oncogenic Factors in Tumor Cells. Biomedicines 2020; 8:103. [PMID: 32365899 PMCID: PMC7277375 DOI: 10.3390/biomedicines8050103] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Garcinol, a polyisoprenylated benzophenone, is the medicinal component obtained from fruits and leaves of Garcinia indica (G. indica) and has traditionally been extensively used for its antioxidant and anti-inflammatory properties. In addition, it has been also been experimentally illustrated to elicit anti-cancer properties. Several in vitro and in vivo studies have illustrated the potential therapeutic efficiency of garcinol in management of different malignancies. It mainly acts as an inhibitor of cellular processes via regulation of transcription factors NF-κB and JAK/STAT3 in tumor cells and have been demonstrated to effectively inhibit growth of malignant cell population. Numerous studies have highlighted the anti-neoplastic potential of garcinol in different oncological transformations including colon cancer, breast cancer, prostate cancer, head and neck cancer, hepatocellular carcinoma, etc. However, use of garcinol is still in its pre-clinical stage and this is mainly attributed to the limitations of conclusive evaluation of pharmacological parameters. This necessitates evaluation of garcinol pharmacokinetics to precisely identify an appropriate dose and route of administration, tolerability, and potency under physiological conditions along with characterization of a therapeutic index. Hence, the research is presently ongoing in the dimension of exploring the precise metabolic mechanism of garcinol. Despite various lacunae, garcinol has presented with promising anti-cancer effects. Hence, this review is motivated by the constantly emerging and promising positive anti-cancerous effects of garcinol. This review is the first effort to summarize the mechanism of action of garcinol in modulation of anti-cancer effect via regulation of different cellular processes.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India;
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia;
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Samruddhi Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur 133001, India;
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
466
|
Larabi A, Barnich N, Nguyen HTT. Emerging Role of Exosomes in Diagnosis and Treatment of Infectious and Inflammatory Bowel Diseases. Cells 2020; 9:cells9051111. [PMID: 32365813 PMCID: PMC7290936 DOI: 10.3390/cells9051111] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
To communicate with each other, cells release exosomes that transfer their composition, including lipids, proteins and nucleic acids, to neighboring cells, thus playing a role in various pathophysiological processes. During an infection with pathogenic bacteria, such as adherent-invasive E. coli (AIEC) associated with Crohn disease, exosomes secreted by infected cells can have an impact on the innate immune responses of surrounding cells to infection. Furthermore, inflammation can be amplified via the exosomal shuttle during infection with pathogenic bacteria, which could contribute to the development of the associated disease. Since these vesicles can be released in various biological fluids, changes in exosomal content may provide a means for the identification of non-invasive biomarkers for infectious and inflammatory bowel diseases. Moreover, evidence suggests that exosomes could be used as vaccines to prime the immune system to recognize and kill invading pathogens, and as therapeutic components relieving intestinal inflammation. Here, we summarize the current knowledge on the role of exosomes in bacterial infections and highlight their potential use as biomarkers, vaccines and conveyers of therapeutic molecules in inflammatory bowel diseases.
Collapse
|
467
|
Yang C, Zhang M, Lama S, Wang L, Merlin D. Natural-lipid nanoparticle-based therapeutic approach to deliver 6-shogaol and its metabolites M2 and M13 to the colon to treat ulcerative colitis. J Control Release 2020; 323:293-310. [PMID: 32335157 DOI: 10.1016/j.jconrel.2020.04.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 01/12/2023]
Abstract
The anti-inflammatory drug candidate, 6-shogaol, has demonstrated excellent efficacies in various in vitro studies. However, its rapid metabolism after oral administration results in poor bioavailability and undetectable in vivo pharmacokinetics. Here, we constructed a natural-lipid (NL) nanoparticle drug delivery system (NP-DDS) to encapsulate 6-shogaol and undertake its controlled release to the proposed drug target (colon). Our in vitro drug-release assay revealed that NL-encapsulated 6-shogaol (6-S-NL) exhibits a delayed drug-release profile compared to free 6-shogaol (free-6-S). Consistent with our expectations, orally administrated 6-S-NL exhibits a superior anti-inflammatory efficacy likely due to the controlled release compared to free 6-S in a dextran sulfate sodium (DSS)-induced mouse model of colitis. Although 6-S-NL treatment yields an enhanced concentration of 6-shogaol at the target site (colon), this concentration is still far below the effective level. We hypothesize that the released 6-shogaol undergoes rapid metabolism and that the metabolites of 6-shogaol may contribute to the anti-inflammatory efficacy of 6-S-NL. We thus examined the in vitro anti-inflammatory efficacies of two highly abundant colonic metabolites, M2 (a cysteine-conjugated metabolite) and M13 (a glutathione-conjugated metabolite), against macrophage cells. Reverse transcription-polymerase chain reaction (RT-PCR) data showed that both M2 and M13 (at 1.0 μg/mL) could down-regulate pro-inflammatory factors (TNF-α, IL-1β, and IL-6) and up-regulate an anti-inflammatory factor (IL-10) in inflamed Raw 264.7 cells. Subsequent in vitro wound-healing assays also confirmed that M2 and M13 accelerate the wound recovery process of Caco-2 cells at the concentrations seen in the colon (1.0 μg/mL). Further, in the DSS-induced mouse model of colitis, oral administration of M2- or M13-loaded NL nanoparticles (M2-NL, M13-NL) demonstrated excellent in vivo wound-healing effects, and these activities were better than those observed for 6-S-NL. Combined with the 6-S-NL's bio-distribution assay, our data show that: the 6-shogaol metabolites, M2 and M13, are more potent anti-inflammatory compounds than 6-shogaol itself; NL nanoparticles can effectively deliver 6-shogaol to the colon, with little accumulation seen in the kidney or liver; and the actions of M2 and M13 mostly confer the anti-inflammatory effect of 6-S-NL. Our results explained the discrepancy between the low tissue concentrations of NL delivered 6-shogaol and its effectiveness against ulcerative colitis (UC) in a mouse model. This study paved the way for further developing the NL-loaded active metabolites, M2 or M13, as novel targeted therapeutic approaches for curing UC.
Collapse
Affiliation(s)
- Chunhua Yang
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA 30303, USA.
| | - Mingzhen Zhang
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA 30303, USA; Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Sudeep Lama
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA 30303, USA
| | - Lixin Wang
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA 30303, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA 30303, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA 30302, USA
| |
Collapse
|
468
|
Functionalization of ginger derived nanoparticles with chitosan to design drug delivery system for controlled release of 5-amino salicylic acid (5-ASA) in treatment of inflammatory bowel diseases: An in vitro study. REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2020.104520] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
469
|
Durán-Lobato M, Niu Z, Alonso MJ. Oral Delivery of Biologics for Precision Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901935. [PMID: 31222910 DOI: 10.1002/adma.201901935] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/02/2019] [Indexed: 05/23/2023]
Abstract
The emerging field of precision medicine is rapidly growing, fostered by the advances in genome mapping and molecular diagnosis. In general, the translation of these advances into precision treatments relies on the use of biological macromolecules, whose structure offers a high specificity and potency. Unfortunately, due to their complex structure and limited ability to overcome biological barriers, these macromolecules need to be administered via injection. The scientific community has devoted significant effort to making the oral administration of macromolecules plausible thanks to the implementation of drug delivery technologies. Here, an overview of the current situation and future prospects in the field of oral delivery of biologics is provided. Technologies in clinical trials, as well as recent and disruptive delivery systems proposed in the literature for local and systemic delivery of biologics including peptides, antibodies, and nucleic acids, are described. Strategies for the specific targeting of gastrointestinal regions-stomach, small bowel, and colon-cell populations, and internalization pathways, are analyzed. Finally, challenges associated with the clinical translation, future prospects, and identified opportunities for advancement in this field are also discussed.
Collapse
Affiliation(s)
- Matilde Durán-Lobato
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Zhigao Niu
- Riddet Institute, Massey University, Palmerston North, 4442, New Zealand
- Food and Bio-based Products Group, AgResearch Ltd, Palmerston North, 4442, New Zealand
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|
470
|
Kalarikkal SP, Prasad D, Kasiappan R, Chaudhari SR, Sundaram GM. A cost-effective polyethylene glycol-based method for the isolation of functional edible nanoparticles from ginger rhizomes. Sci Rep 2020; 10:4456. [PMID: 32157137 PMCID: PMC7064537 DOI: 10.1038/s41598-020-61358-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/26/2020] [Indexed: 12/18/2022] Open
Abstract
Edible nanoparticles (ENPs) are nano-sized vesicles derived from edible plants. These ENPs are loaded with plant derived microRNAs, protein, lipids and phytochemicals. Recently, ginger derived ENPs was shown to prevent inflammatory bowel diseases and colon cancer, in vivo, highlighting their therapeutic potential. Conventionally, differential centrifugation with an ultra-centrifugation step is employed to purify these ENPs which imposes limitation on the cost-effectiveness of their purification. Herein, we developed polyethylene glycol-6000 (PEG6000) based ginger ENP purification (PEG-ENPs) method, which eliminates the need for expensive ultracentrifugation. Using different PEG6000 concentrations, we could recover between 60% to 90% of ENPs compared to ultracentrifugation method. PEG-ENPs exhibit near identical size and zeta potential similar to ultra-ENPs. The biochemical composition of PEG-ENPs, such as proteins, lipids, small RNAs and bioactive content is comparable to that of ultra-ENPs. In addition, similar to ultra-ENPs, PEG-ENPs are efficiently taken up by the murine macrophages and protects cells from hydrogen peroxide induced oxidative stress. Since PEG has been approved as food additive, the PEG method described here will provide a cost-effective alternative to purify ENPs, which can be directly used as a dietary supplement in therapeutic formulations.
Collapse
Affiliation(s)
- Sreeram Peringattu Kalarikkal
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CFTRI Campus, Mysuru, Karnataka, India
- Department of Biochemistry, CSIR-CFTRI, Mysuru, Karnataka, India
| | - Durga Prasad
- Department of Spice & Flavor Science, CSIR-CFTRI, Mysuru, Karnataka, India
| | - Ravi Kasiappan
- Department of Biochemistry, CSIR-CFTRI, Mysuru, Karnataka, India
| | - Sachin R Chaudhari
- Department of Spice & Flavor Science, CSIR-CFTRI, Mysuru, Karnataka, India
| | - Gopinath M Sundaram
- Academy of Scientific and Innovative Research (AcSIR), CSIR-CFTRI Campus, Mysuru, Karnataka, India.
- Department of Biochemistry, CSIR-CFTRI, Mysuru, Karnataka, India.
| |
Collapse
|
471
|
Liu B, Lu Y, Chen X, Muthuraj PG, Li X, Pattabiraman M, Zempleni J, Kachman SD, Natarajan SK, Yu J. Protective Role of Shiitake Mushroom-Derived Exosome-Like Nanoparticles in D-Galactosamine and Lipopolysaccharide-Induced Acute Liver Injury in Mice. Nutrients 2020; 12:nu12020477. [PMID: 32069862 PMCID: PMC7071144 DOI: 10.3390/nu12020477] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Fulminant hepatic failure (FHF) is a rare, life-threatening liver disease with a poor prognosis. Administration of D-galactosamine (GalN) and lipopolysaccharide (LPS) triggers acute liver injury in mice, simulating many clinical features of FHF in humans; therefore, this disease model is often used to investigate potential therapeutic interventions to treat FHF. Recently, suppression of the nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome, was shown to alleviate the severity of GalN/LPS-induced liver damage in mice. Therefore, the goal of this study was to find dietary exosome-like nanoparticles (ELNs) with therapeutic potential in curbing FHF by suppressing the NLRP3 inflammasome. Seven commonly consumed mushrooms were used to extract ELNs. These mushrooms were found to contain ELNs composed of RNAs, proteins, and lipids. Among these mushroom-derived ELNs, only shiitake mushroom-derived ELNs (S-ELNs) substantially inhibited NLRP3 inflammasome activation by preventing inflammasome formation in primary macrophages. S-ELNs also suppressed the secretion of interleukin (IL)-6, as well as both protein and mRNA levels of the Il1b gene. Remarkably, pre-treatment with S-ELNs protected mice from GalN/LPS-induced acute liver injury. Therefore, S-ELNs, identified as potent new inhibitors of the NLRP3 inflammasome, represent a promising class of agents with the potential to combat FHF.
Collapse
Affiliation(s)
- Baolong Liu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Yizhu Lu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Xingyi Chen
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Xingzhi Li
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Mahesh Pattabiraman
- Department of Chemistry, University of Nebraska-Kearney, Kearney, NE 68849, USA;
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Stephen D. Kachman
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (B.L.); (Y.L.); (X.C.)
- Correspondence: ; Tel.: +1-402-472-7013
| |
Collapse
|
472
|
Advances in colon-targeted nano-drug delivery systems: challenges and solutions. Arch Pharm Res 2020; 43:153-169. [DOI: 10.1007/s12272-020-01219-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
|
473
|
Sung J, Yang C, Collins JF, Merlin D. Preparation and Characterization of Ginger Lipid-derived Nanoparticles for Colon-targeted siRNA Delivery. Bio Protoc 2020; 10:e3685. [PMID: 32782915 DOI: 10.21769/bioprotoc.3685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Synthetic nanoparticle-based drug delivery system is widely known for its ability to increase the efficacy and specificity of loaded drugs, but it often suffers from relatively higher immunotoxicity and higher costs as compared to traditional drug formulations. Contrarily, plant-derived nanoparticles appear to be free from these limitations of synthetic nanoparticles; they are naturally occurring biocompatible vesicles that do not generate immunotoxicity and are easy to obtain. Additionally, lipids isolated from plant-derived nanoparticles have shown the capability of assembling themselves to spherical nano-sized liposomal particles. Herein, we employ lipids extracted from ginger-derived nanoparticles and load them with therapeutic siRNA (CD98-siRNA) to create CD98-siRNA/ginger-lipid nanoparticles. Characterization of the CD98-siRNA/ginger-lipid nanoparticles showed that they present a spherical shape, with a diameter of around 189.5 nm. The surface zeta potential of the nanoparticles varies from -18.1 to -18.4 mV. Furthermore, in recent research, the CD98-siRNA/ginger-lipid nanoparticles have shown specific colon targeting capability and excellent anti-inflammatory efficacy in a Dextran Sodium Sulfate (DSS) induced mouse model of colitis.
Collapse
Affiliation(s)
- Junsik Sung
- Institute for Biomedical Science, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Chunhua Yang
- Institute for Biomedical Science, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Didier Merlin
- Institute for Biomedical Science, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
474
|
Maheswari P, Ponnusamy S, Harish S, Ganesh M, Hayakawa Y. Hydrothermal synthesis of pure and bio modified TiO2: Characterization, evaluation of antibacterial activity against gram positive and gram negative bacteria and anticancer activity against KB Oral cancer cell line. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2018.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
475
|
Yang M, Zhang F, Yang C, Wang L, Sung J, Garg P, Zhang M, Merlin D. Oral Targeted Delivery by Nanoparticles Enhances Efficacy of an Hsp90 Inhibitor by Reducing Systemic Exposure in Murine Models of Colitis and Colitis-Associated Cancer. J Crohns Colitis 2020; 14:130-141. [PMID: 31168612 DOI: 10.1093/ecco-jcc/jjz113] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Heat shock protein 90 [Hsp90]-targeted therapy has been proposed as a promising strategy for the treatment of ulcerative colitis [UC] and colitis-associated cancer [CAC]. Systemic administration of the Hsp90 inhibitor, 17-AAG, was found to be profoundly protective in preclinical mouse models of inflammatory bowel disease [IBD]. However, the therapeutic potential of 17-AAG is limited by potential side effects associated with its systemic exposure and the modest bioavailability afforded by its oral administration. METHODS To address these issues, we used a versatile single-step surface-functionalizing technique to prepare a 17-AAG oral delivery system using PLGA/PLA-PEG-FA nanoparticles [NP-PEG-FA/17-AAG]. RESULTS NP-PEG-FA could be efficiently taken up by mouse Colon-26 cells and activated Raw 264.7 cells in vitro and by inflamed mouse colitis tissues in vivo. The therapeutic efficacy of orally administrated NP-PEG-FA/17-AAG was evaluated in in vivo models using dextran sulphate sodium [DSS]-induced UC and azoxymethane [AOM]/DSS-induced CAC, and the results indicated that NP-PEG-FA/17-AAG significantly alleviated the symptoms of UC and CAC. More importantly, our inflamed colitis-targeted 17-AAG nano-formulation reduced systemic exposure and provided a degree of therapeutic response similar to that obtained by systemic administration [intraperitoneal] of 17-AAG, but at a ten-fold lower dose. CONCLUSIONS We describe a convenient, orally administrated 17-AAG delivery system that exhibits enhanced efficacy in UC and CAC therapy while reducing systemic exposure. This system may represent a promising therapeutic approach for treating UC and CAC.
Collapse
Affiliation(s)
- Mei Yang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Fang Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Chunhua Yang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Lixin Wang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Junsik Sung
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Pallavi Garg
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Mingzhen Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
476
|
Gupta J, Sharma S, Sharma NR, Kabra D. Phytochemicals enriched in spices: a source of natural epigenetic therapy. Arch Pharm Res 2019; 43:171-186. [DOI: 10.1007/s12272-019-01203-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023]
|
477
|
Zhang M, Yang C, Yan X, Sung J, Garg P, Merlin D. Highly Biocompatible Functionalized Layer-by-Layer Ginger Lipid Nano Vectors Targeting P-selectin for Delivery of Doxorubicin to Treat Colon Cancer. ADVANCED THERAPEUTICS 2019; 2:1900129. [PMID: 33043129 PMCID: PMC7546358 DOI: 10.1002/adtp.201900129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Indexed: 12/15/2022]
Abstract
A biocompatible natural nanoparticle drug delivery system that has specific cancer-targeting function holds vast promise for cancer therapy. Here, a fucoidan/poly-lysine-functionalized layer-by-layer ginger-derived lipid vector (LbL-GDLV) was designed to target P-selectin (overexpressed by endothelial cells) and deliver a loaded drug into vascularized colon cancer. In vitro, LbL-GDLVs selectively bound to P-selectin, and the degradation of the fucoidan outer layer in a milieu similar to the cancer microenvironment resulted in rapid attachment of the cancer cell and internalization of the remaining positively charged poly-lysine coated-GDLVs. Upon enzymolysis of the poly-lysine layer inside the cancer cell, the GDLV core released loaded doxorubicin (Dox) which had the expected effects. In vivo bio-distribution studies showed that intravenously injected LbL-GDLVs exhibited enhanced accumulation at the vascularized tumor site (~ 4.4-fold higher than control vesicles), presumably due to P-selectin-mediated targeting plus the enhanced permeability and retention effect (EPR). In two animal models used to screen anti-cancer efficacy (Luc-HT-29 and HCT-116 xenografts), Dox-loaded LbL-GDLVs (LbL-GDLVs/Dox) significantly inhibited tumor growth and demonstrated much better therapeutic efficiency than free Dox. More importantly, LbL-GDLVs/Dox exhibited excellent biocompatibility, and LbL-GDLVs encapsulation largely reduced the cardiotoxicity of free Dox and avoided the notorious drug resistance of colon cells against free Dox. Together, these findings demonstrate the potential of our newly designed and highly biocompatible plant-derived LbL nanoparticles and their precise colon cancer drug delivery function.
Collapse
Affiliation(s)
- Mingzhen Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, 30302, United States
| | - Chunhua Yang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, 30302, United States
| | - Xiangji Yan
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Junsik Sung
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, 30302, United States
| | - Pallavi Garg
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, 30302, United States
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, 30302, United States
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, 30033, United States
| |
Collapse
|
478
|
Yang C, Merlin D. Can naturally occurring nanoparticle-based targeted drug delivery effectively treat inflammatory bowel disease? Expert Opin Drug Deliv 2019; 17:1-4. [PMID: 31770040 DOI: 10.1080/17425247.2020.1698543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Chunhua Yang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| |
Collapse
|
479
|
Phytofabrication of Nanoparticles as Novel Drugs for Anticancer Applications. Molecules 2019; 24:molecules24234246. [PMID: 31766544 PMCID: PMC6930546 DOI: 10.3390/molecules24234246] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/30/2019] [Accepted: 11/08/2019] [Indexed: 02/05/2023] Open
Abstract
Cancer is one of the foremost causes of death globally and also the major stumbling block of increasing life expectancy. Although the primary treatment of surgical resection, chemotherapy, and radiotherapy have greatly reduced the mortality of cancer, the survival rate is still low because of the metastasis of tumor, a range of adverse drug reactions, and drug resistance. For all this, it is relevant to mention that a growing amount of research has shown the anticarcinogenic effect of phytochemicals which can modulate the molecular pathways and cellular events include apoptosis, cell proliferation, migration, and invasion. However, their pharmacological potential is hindered by their low water solubility, low stability, poor absorption, and rapid metabolism. In this scenario, the development of nanotechnology has created novel formulations to maximize the potential use of phytochemicals in anticancer treatment. Nanocarriers can enhance the solubility and stability of phytochemicals, prolong their half-life in blood and even achieve site-targeting delivery. This review summarizes the advances in utilizing nanoparticles in cancer therapy. In particular, we introduce several applications of nanoparticles combined with apigenin, resveratrol, curcumin, epigallocatechin-3-gallate, 6-gingerol, and quercetin in cancer treatment.
Collapse
|
480
|
Thrifty, Rapid Intestinal Monolayers (TRIM) Using Caco-2 Epithelial Cells for Oral Drug Delivery Experiments. Pharm Res 2019; 36:172. [PMID: 31659456 DOI: 10.1007/s11095-019-2712-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/30/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE Caco-2 monolayers are the most common model of the intestinal epithelium and are critical to the development of oral drug delivery strategies and gastrointestinal disease treatments. However, current monolayer systems are cost- and/or time-intensive, hampering progress. This study evaluates two separate methods to reduce resource input: FB Essence as a fetal bovine serum (FBS) alternative and a new, 3-day Caco-2 system deemed "thrifty, rapid intestinal monolayers" (TRIM). METHODS Caco-2 cells were cultured with FB Essence and compared to cells in 10% FBS for proliferation and monolayer formation. TRIM were compared to commonly-used 21-day and Corning® HTS monolayer systems, as well as mouse intestines, for permeability behavior, epithelial gene expression, and tight junction arrangement. RESULTS No amount of FB Essence maintained Caco-2 cells beyond 10 passages. In contrast, TRIM compared favorably in permeability and gene expression to intestinal tissues. Furthermore, TRIM cost $109 and required 1.3 h of time per 24-well plate, compared to $164 and 3.7 h for 21-day monolayers, and $340 plus 1.0 h for the HTS system. CONCLUSIONS TRIM offer a new approach to generating Caco-2 monolayers that resemble the intestinal epithelium. They are anticipated to accelerate the pace of in vitro intestinal experiments while easing financial burden.
Collapse
|
481
|
Ma Y, Tong X, Huang Y, Zhou X, Yang C, Chen J, Dai F, Xiao B. Oral Administration of Hydrogel-Embedding Silk Sericin Alleviates Ulcerative Colitis through Wound Healing, Anti-Inflammation, and Anti-Oxidation. ACS Biomater Sci Eng 2019; 5:6231-6242. [DOI: 10.1021/acsbiomaterials.9b00862] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | | | - Chunhua Yang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, 100 Piedmont Avenue SE, Atlanta, Georgia 30302, United States
| | | | | | - Bo Xiao
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, 100 Piedmont Avenue SE, Atlanta, Georgia 30302, United States
| |
Collapse
|
482
|
|
483
|
Lin Q, Qu M, Zhou B, Patra HK, Sun Z, Luo Q, Yang W, Wu Y, Zhang Y, Li L, Deng L, Wang L, Gong T, He Q, Zhang L, Sun X, Zhang Z. Exosome-like nanoplatform modified with targeting ligand improves anti-cancer and anti-inflammation effects of imperialine. J Control Release 2019; 311-312:104-116. [PMID: 31484040 DOI: 10.1016/j.jconrel.2019.08.037] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 12/20/2022]
Abstract
Currently, most anti-cancer therapies are still haunted by serious and deleterious adverse effects. Here, we report a highly biocompatible tumor cell-targeting delivery systems utilizing exosome-like vesicles (ELVs) that delivers a low-toxicity anti-cancer agent imperialine against non-small cell lung cancer (NSCLC). First, we introduced a novel micelle-aided method to efficiently load imperialine into intact ELVs. Then, integrin α3β1-binding octapeptide cNGQGEQc was modified onto ELV platform for tumor targeting as integrin α3β1 is overexpressed on NSCLC cells. This system not only significantly improved imperialine tumor accumulation and retention, but also had extremely low systemic toxicity both in vitro and in vivo. Our discoveries offer new ways to utilize ELV more efficiently for both drug loading and targeting. The solid pharmacokinetics improvement and extraordinary safety of this system also highlight possibilities of alternative long course cancer therapies using similar strategies.
Collapse
Affiliation(s)
- Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Mengke Qu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Bingjie Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Hirak K Patra
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK; Department of Clinical and Experimental Medicine, Linkoping University, Linkoping 58185, Sweden; Wolfson College, University of Cambridge, Cambridge CB3 9BB, United Kingdom
| | - Zihan Sun
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Qiong Luo
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Wenyu Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Yongcui Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Yu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Lin Li
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Lang Deng
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Leilei Wang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| |
Collapse
|
484
|
Strategies for the use of Extracellular Vesicles for the Delivery of Therapeutics. J Neuroimmune Pharmacol 2019; 15:422-442. [PMID: 31456107 DOI: 10.1007/s11481-019-09873-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are nanosized, membrane-bound vesicles released from eukaryotic and prokaryotic cells that can transport cargo containing DNA, RNA, lipids and proteins, between cells as a means of intercellular communication. Although EVs were initially considered to be cellular debris deprived of any essential biological functions, emerging literature highlights the critical roles of EVs in the context of intercellular signaling, maintenance of tissue homeostasis, modulation of immune responses, inflammation, cancer progression, angiogenesis, and coagulation under both physiological and pathological states. Based on the ability of EVs to shuttle proteins, lipids, carbohydrates, mRNAs, long non-coding RNAs (lncRNAs), microRNAs, chromosomal DNA, and mitochondrial DNA into target cells, the presence and content of EVs in biofluids have been exploited for biomarker research in the context of diagnosis, prognosis and treatment strategies. Additionally, owing to the characteristics of EVs such as stability in circulation, biocompatibility as well as low immunogenicity and toxicity, these vesicles have become attractive systems for the delivery of therapeutics. More recently, EVs are increasingly being exploited as conduits for delivery of therapeutics for anticancer strategies, immunomodulation, targeted drug delivery, tissue regeneration, and vaccination. In this review, we highlight and discuss the multiple strategies that are employed for the use of EVs as delivery vehicles for therapeutic agents, including the potential advantages and challenges involved. Graphical abstract.
Collapse
|
485
|
Puri D, Khatik GL, Shunmugaperumal T. Studies on olive-and silicone-oils-based Janus macroemulsions containing ginger to manage primary dysmenorrheal pain. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:276-285. [PMID: 30948062 DOI: 10.1016/j.msec.2019.01.137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 11/28/2022]
Abstract
Ginger (GIN) powder-loaded oil-in-water (o/w) macroemulsions were prepared based on olive-and silicone-oils. The dispersed oil droplets with paired-beans structure were evident and thus the final emulsion can be termed as Janus macroemulsions. The objectives of the present study are (1) to identify the marker compound present in GIN powder via HPLC analysis, (2) to process the GIN powder via anti-solvent precipitation technique, (3) to see the solubility of GIN powder in various single oils or oil combination, (4) to optimize the GIN-loaded o/w macroemulsions using the central composite design (CCD) with respect to mean particle size of dispersed oil droplets and highest percentage drug entrapment efficiency values (DEE) and (5) to evaluate the pain reducing activity of optimized GIN-loaded macroemulsion via in vivo primary dysmenorrhea (PD) mice model. Both predicted and obtained values of percentage DEE (76.29 Vs.76.09) and mean particle size (245.99 Vs. 272.51 μm) were almost the same indicating the CCD statistical design applicability. The optimized Janus macroemulsion was stable at 4 °C for over a period of 90 days. Using the PD mice model, the counting of writhing reaction produced by the tested GIN-loaded macroemulsions at low and high doses did not reveal significant difference in comparison to the positive control (aspirin treated). Only the high dose of GIN-loaded macroemulsion was able to restore the uterine tissue's normal histomorphological structure after the H & E staining. Nevertheless, the paired beans structure should be tested for entrapping the plant-derived drugs having dissimilar physicochemical characteristics but similar therapeutic activity.
Collapse
Affiliation(s)
- Diksha Puri
- Lovely School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T.Road (NH-1), Phagwara, Jalandhar, Punjab 144411, India
| | - Gopal Lal Khatik
- Lovely School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T.Road (NH-1), Phagwara, Jalandhar, Punjab 144411, India
| | - Tamilvanan Shunmugaperumal
- Lovely School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T.Road (NH-1), Phagwara, Jalandhar, Punjab 144411, India; National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, C/O NETES Institute of Technology & Science, NH-37, Santipur, Parli Part, Mirza, Assam 781125, India.
| |
Collapse
|
486
|
Gao C, Liu L, Zhou Y, Bian Z, Wang S, Wang Y. Novel drug delivery systems of Chinese medicine for the treatment of inflammatory bowel disease. Chin Med 2019; 14:23. [PMID: 31236131 PMCID: PMC6580650 DOI: 10.1186/s13020-019-0245-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory disease that comprises ulcerative colitis (UC) and Crohn's disease (CD). IBD involves the ileum, rectum, and colon, and common clinical manifestations of IBD are diarrhea, abdominal pain, and even bloody stools. Currently, non-steroidal anti-inflammatory drugs, glucocorticoids, and immunosuppressive agents are used for the treatment of IBD, while their clinical application is severely limited due to unwanted side effects. Chinese medicine (CM) is appealing more and more attention and investigation for the treatment of IBD owing to the potent anti-inflammation pharmacological efficacy and high acceptance by patients. In recent years, novel drug delivery systems are introduced apace to encapsulate CM and many CM-derived active constituents in order to improve solubility, stability and targeting ability. In this review, advanced drug delivery systems developed in the past and present to deliver CM for the treatment of IBD are summarized and future directions are discussed.
Collapse
Affiliation(s)
- Caifang Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, SAR China
| | - Lijuan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, SAR China
- PU-UM Innovative Institute of Chinese Medical Sciences, Guangdong-Macau Traditional Chinese Medicine Technology Industrial Park Development Co., Ltd, Hengqin New Area, Zhuhai, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan China
| | - Yangyang Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, SAR China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, SAR China
- Leiden University European Center for Chinese Medicine and Natural Compounds, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, SAR China
| |
Collapse
|
487
|
Dubey KK, Luke GA, Knox C, Kumar P, Pletschke BI, Singh PK, Shukla P. Vaccine and antibody production in plants: developments and computational tools. Brief Funct Genomics 2019; 17:295-307. [PMID: 29982427 DOI: 10.1093/bfgp/ely020] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Plants as bioreactors have been widely used to express efficient vaccine antigens against viral, bacterial and protozoan infections. To date, many different plant-based expression systems have been analyzed, with a growing preference for transient expression systems. Antibody expression in diverse plant species for therapeutic applications is well known, and this review provides an overview of various aspects of plant-based biopharmaceutical production. Here, we highlight conventional and gene expression technologies in plants along with some illustrative examples. In addition, the portfolio of products that are being produced and how they relate to the success of this field are discussed. Stable and transient gene expression in plants, agrofiltration and virus infection vectors are also reviewed. Further, the present report draws attention to antibody epitope prediction using computational tools, one of the crucial steps of vaccine design. Finally, regulatory issues, biosafety and public perception of this technology are also discussed.
Collapse
Affiliation(s)
- Kashyap Kumar Dubey
- Department of Biotechnology, Central University of Haryana, Jant-Pali Mahendergarh, Haryana, India.,Microbial Process Development Laboratory, University Institute of Engineering and Technology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Garry A Luke
- Centre for Biomolecular Sciences, School of Biology, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, Scotland
| | - Caroline Knox
- Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Punit Kumar
- Microbial Process Development Laboratory, University Institute of Engineering and Technology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Brett I Pletschke
- Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Puneet Kumar Singh
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
488
|
Abstract
Identification of active constituents of our diet is crucial to understand the impact of food on health, and disease development, and for the formulation of functional food and nutraceuticals. Until now research into the pharmacological properties of the components of our diet has focused on vitamins, sterols, polyphenols, fiber, etc. But very recently, it has been found that plants contain various types of vesicles which are in contact with the intestinal tract throughout our lives. They participate in intestinal tissue renewal processes and modulate gut microbiota in healthy subjects and have important biological functions against inflammatory diseases (e.g.; colitis injury, liver steatosis) or cancers associated with their specific lipid and miRNA content. In addition, recent data have suggested that plant-derived nanovesicles would be excellent candidates for the delivery of therapeutic agents (e.g.; anti-cancerous drugs, siRNAs) or poorly soluble natural compounds (e.g.; curcumin), as they are able to cross mammalian barriers without inducing either an inflammatory response or necrosis, conversely to conventional liposomes. It is thus important to consider these plant-derived vesicles as new components of our food in order to evaluate their potential for health benefit and food-derived technology.
Collapse
Affiliation(s)
- Sophie Rome
- CarMeN Laboratory (UMR INSERM 1060-INRA 1397, INSA), Lyon-Sud Faculty of Medicine, University of Lyon, 69310-Pierre-Bénite, France.
| |
Collapse
|
489
|
Abstract
The nucleotide-binding domain and leucine-rich repeat-containing family, pyrin domain-containing 3 (NLRP3) inflammasome is a key regulator of innate immune responses, and its aberrant activation is implicated in the pathogenesis of many diseases such as Alzheimer's disease and type 2 diabetes. Targeting the NLRP3 inflammasome could hold promise to combat these complex diseases, but therapies specifically inhibiting the NLRP3 inflammasome have not been developed for patient treatment. The current study aimed to identify food-borne exosome-like nanoparticles (ELNs) that inhibit NLRP3 inflammasome activity. Nine vegetables or fruits were selected to extract ELNs, which were examined for their inhibitory effects on activation of the NLRP3 inflammasome in primary macrophages. Although most of the tested ELNs posed minimal impacts, the ELNs from ginger rhizomes (G-ELNs) strongly inhibited NLRP3 inflammasome activation. The G-ELNs contained lipids, proteins, and RNAs and were easily taken up by macrophages. G-ELN treatment suppressed pathways downstream of inflammasome activation including caspase1 autocleavage, interleukin (IL)-1β and IL-18 secretion, and pyroptotic cell death. Apoptotic speck protein containing a caspase recruitment domain (ASC) oligomerization and speck formation assays indicated that G-ELNs blocked assembly of the NLRP3 inflammasome. The lipids in G-ELNs, rather than the RNAs or proteins, were responsible for the inhibitory activity observed. Together, the data suggested G-ELNs as new potent agents that block NLRP3 inflammasome assembly and activation. The unique features of G-ELNs including biomolecule protection and tissue bioavailability should facilitate the development of G-ELN-based therapy to target the NLRP3 inflammasome in the disease settings.
Collapse
Affiliation(s)
- Xingyi Chen
- Department of Nutrition and Health Sciences , University of Nebraska Lincoln , 230 Filley Hall , Lincoln , Nebraska 68583-0922 , United States
| | - You Zhou
- Center for Biotechnology , University of Nebraska Lincoln , E117 Beadle Center, Lincoln , Nebraska 68588-0665 , United States
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences , University of Nebraska Lincoln , 230 Filley Hall , Lincoln , Nebraska 68583-0922 , United States
| |
Collapse
|
490
|
Sundaram GM. Dietary non-coding RNAs from plants: Fairy tale or treasure? Noncoding RNA Res 2019; 4:63-68. [PMID: 31193509 PMCID: PMC6533053 DOI: 10.1016/j.ncrna.2019.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/26/2018] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
The past two decades have witnessed soaring interest in the field of non-coding RNAs, largely attributed by its regulatory role in controlling two third of human transcriptional output. Though, there are several classes of non-coding RNAs found in nature, microRNAs takes the central stage because of their pleiotropic roles. In particular, extracellular microRNAs are gaining traction due to their relative stability and bio availability. Extracellular microRNAs has been shown to occur in all living organisms, including dietary plants. Some of the recent reports suggest that these dietary microRNAs pass through the gut, enter systemic circulation and exert biological effects on animal physiology. However, evidences against this hypothesis are also presented in literature and hence this area has been strongly debated. In this review, I will briefly summarise the evidences accumulated for and against this hypothesis and discuss potential implications of such findings in human health.
Collapse
Affiliation(s)
- Gopinath M. Sundaram
- Department of Biochemistry, Central Food Technological Research Institute, Mysore, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
491
|
Mao QQ, Xu XY, Cao SY, Gan RY, Corke H, Beta T, Li HB. Bioactive Compounds and Bioactivities of Ginger ( Zingiber officinale Roscoe). Foods 2019; 8:E185. [PMID: 31151279 PMCID: PMC6616534 DOI: 10.3390/foods8060185] [Citation(s) in RCA: 463] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 05/26/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Ginger (Zingiber officinale Roscoe) is a common and widely used spice. It is rich in various chemical constituents, including phenolic compounds, terpenes, polysaccharides, lipids, organic acids, and raw fibers. The health benefits of ginger are mainly attributed to its phenolic compounds, such as gingerols and shogaols. Accumulated investigations have demonstrated that ginger possesses multiple biological activities, including antioxidant, anti-inflammatory, antimicrobial, anticancer, neuroprotective, cardiovascular protective, respiratory protective, antiobesity, antidiabetic, antinausea, and antiemetic activities. In this review, we summarize current knowledge about the bioactive compounds and bioactivities of ginger, and the mechanisms of action are also discussed. We hope that this updated review paper will attract more attention to ginger and its further applications, including its potential to be developed into functional foods or nutraceuticals for the prevention and management of chronic diseases.
Collapse
Affiliation(s)
- Qian-Qian Mao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Xiao-Yu Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Shi-Yu Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ren-You Gan
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Harold Corke
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Trust Beta
- Department of Food & Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
492
|
Akuma P, Okagu OD, Udenigwe CC. Naturally Occurring Exosome Vesicles as Potential Delivery Vehicle for Bioactive Compounds. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2019. [DOI: 10.3389/fsufs.2019.00023] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
493
|
Nikkhah-Bodaghi M, Maleki I, Agah S, Hekmatdoost A. Zingiber officinale and oxidative stress in patients with ulcerative colitis: A randomized, placebo-controlled, clinical trial. Complement Ther Med 2019; 43:1-6. [PMID: 30935515 DOI: 10.1016/j.ctim.2018.12.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/22/2018] [Accepted: 12/29/2018] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Oxidative stress plays an essential role in ulcerative colitis (UC) initiation and severity. We aimed to investigate the effect of ginger as a well-known antioxidant agent on the quality of life, disease activity index and oxidative stress in patients with UC. METHODS Forty six patients with active mild to moderate UC randomly assigned to consume 2000 mg/day dried ginger powder in 4 capsules or similar placebo capsules for 12 weeks. Disease activity index, quality of life and some oxidative stress factors were measured before, at the middle and at the end of the intervention through valid and reliable questionnaires and blood sampling. RESULTS Ginger reduced Malondialdehyde (MDA) significantly after 6 weeks (p = 0.003) and 12 weeks (p < 0.001) of intervention, whereas it did not affect serum total anti-oxidant capacity (TAC). The scores of severity of disease activity at 12th week was significantly improved in ginger group in comparison to placebo (p = 0.017). Moreover, ginger increased patients quality of life significantly at 12th week (p = 0.039). CONCLUSION Our data indicate that ginger supplementation can improve treatment of patients with UC. Further clinical trials with different dosages and duration of ginger or its standard extract supplementation are needed to obtain firm conclusion.
Collapse
Affiliation(s)
- Mehrnaz Nikkhah-Bodaghi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Iradj Maleki
- Gut and Liver Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Shahram Agah
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
494
|
Ulcerative colitis-specific delivery of keratinocyte growth factor by neutrophils-simulated liposomes facilitates the morphologic and functional recovery of the damaged colon through alleviating the inflammation. J Control Release 2019; 299:90-106. [DOI: 10.1016/j.jconrel.2019.02.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/06/2019] [Accepted: 02/22/2019] [Indexed: 01/23/2023]
|
495
|
Wang X, Zhang M, Flores SRL, Woloshun RR, Yang C, Yin L, Xiang P, Xu X, Garrick MD, Vidyasagar S, Merlin D, Collins JF. Oral Gavage of Ginger Nanoparticle-Derived Lipid Vectors Carrying Dmt1 siRNA Blunts Iron Loading in Murine Hereditary Hemochromatosis. Mol Ther 2019; 27:493-506. [PMID: 30713087 PMCID: PMC6401192 DOI: 10.1016/j.ymthe.2019.01.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles (NPs) have been utilized to deliver drugs to the intestinal epithelium in vivo. Moreover, NPs derived from edible plants are less toxic than synthetic NPs. Here, we utilized ginger NP-derived lipid vectors (GDLVs) in a proof-of-concept investigation to test the hypothesis that inhibiting expression of divalent metal-ion transporter 1 (Dmt1) would attenuate iron loading in a mouse model of hereditary hemochromatosis (HH). Initial experiments using duodenal epithelial organ cultures from intestine-specific Dmt1 knockout (KO) (Dmt1int/int) mice in the Ussing chamber established that Dmt1 is the only active iron importer during iron-deficiency anemia. Further, when Dmt1int/int mice were crossed with mice lacking the iron-regulatory hormone, hepcidin (Hepc-/-), iron loading was abolished. Hence, intestinal Dmt1 is required for the excessive iron absorption that typifies HH. Additional experiments established a protocol to produce GDLVs carrying functional Dmt1 small interfering RNAs (siRNAs) and to target these gene delivery vehicles to the duodenal epithelium in vivo (by incorporating folic acid [FA]). When FA-GDLVs carrying Dmt1 siRNA were administered to weanling Hepc-/- mice for 16 days, intestinal Dmt1 mRNA expression was attenuated and tissue iron accumulation was blunted. Oral delivery of functional siRNAs by FA-GDLVs is a suitable therapeutic approach to mitigate iron loading in murine HH.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Mingzhen Zhang
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Shireen R L Flores
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Regina R Woloshun
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Chunhua Yang
- Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Liangjie Yin
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Ping Xiang
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA; State Key Lab of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Xiaodong Xu
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Michael D Garrick
- Department of Biochemistry, State University of New York (SUNY), Buffalo, NY, USA
| | | | - Didier Merlin
- Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
496
|
CTGF-mediated ERK signaling pathway influences the inflammatory factors and intestinal flora in ulcerative colitis. Biomed Pharmacother 2019; 111:1429-1437. [DOI: 10.1016/j.biopha.2018.12.063] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/28/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
|
497
|
Cao J, Cheng J, Xi S, Qi X, Shen S, Ge Y. Alginate/chitosan microcapsules for in-situ delivery of the protein, interleukin-1 receptor antagonist (IL-1Ra), for the treatment of dextran sulfate sodium (DSS)-induced colitis in a mouse model. Eur J Pharm Biopharm 2019; 137:112-121. [PMID: 30779979 DOI: 10.1016/j.ejpb.2019.02.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/25/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022]
Abstract
Targeted delivery of bioactive compounds such as proteins to the colon has numerous advantages for the therapeutic treatment of inflammatory bowel disease. The present study sought to fabricate alginate/chitosan microcapsules containing IL-1Ra (Alg/Chi/IL-1Ra MC) via a single-step electrospraying method. Two important factors of efficacy were measured-the pH-responsiveness of the microcapsule and the in-vitro drug release profile. The DSS-induced colitis mouse model was used to evaluate the therapeutic effect of the Alg/Chi/IL-1Ra microcapsules, with results showing the protective effect of the Alg/Chi microcapsules for the passage of IL-1Ra through the harsh environment of the upper gastrointestinal tract. This effect was owing to the pH-sensitive response of the microcapsule, which allowed the targeted release of IL-1Ra in the colon. DAI evaluation, colon length, colon tissue morphology, histologic damage scores and relative protein concentrations (MPO, TNF-α and IL-1β) demonstrated that the Alg/Chi/IL-1Ra microcapsules alleviated DSS-induced colitis in mice. The present study thus demonstrates a practical means of oral delivery of proteins, in-situ colon release, and a promising application of IL-1Ra in the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Jin Cao
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China
| | - Jin Cheng
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China
| | - Siyu Xi
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China
| | - Song Shen
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China
| | - Yanru Ge
- School of Pharmacy, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu, China.
| |
Collapse
|
498
|
Kumar S, Sharma B, Thakur K, Bhardwaj TR, Prasad DN, Singh RK. Recent Advances in the Development of Polymeric Nanocarrier Formulations for the Treatment of Colon Cancer. DRUG DELIVERY LETTERS 2019; 9:2-14. [DOI: 10.2174/2210303108666181109120710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 01/04/2025]
Abstract
Background:
Many efforts have been explored in the last decade to treat colon cancer but
nanoparticulate drug delivery systems are making a vital contribution in the improvement of drug delivery
to colon cancer cells.
Objective:
In this review, we attempt to highlight recent advancements in the development of novel
drug delivery systems of nanoparticles for the targeted drug delivery to colon. Polymers like Epithelial
Cell Adhesion Molecule (EpCAM) aptamer chitosan, Hyaluronic Acid (HA), Chitosan (CS)–
Carboxymethyl Starch (CMS), silsesquioxane capped mesoporous silica, Near IR (NIR) fluorescent
Human Serum Albumin (HAS), poly(ethylene glycol)-conjugated hyaluronic acid etc. have been discussed
by employing various anticancer drugs like doxorubicin, oxaliplatin, paclitaxel, 5-fluorouracil
etc.
Conclusion:
These novel drug delivery systems have been determined to be more efficacious in terms
of stability, sustained and targeted drug delivery, therapeutic efficacy, improved bioavailability and enhanced
anticancer activity.
Collapse
Affiliation(s)
- Sahil Kumar
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Sohna, Dist. Gurugram- 122103, Haryana, India
| | - Bandna Sharma
- School of Pharmacy & Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi-173205, India
| | - Kiran Thakur
- School of Pharmacy & Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi-173205, India
| | - Tilak R. Bhardwaj
- School of Pharmacy & Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi-173205, India
| | - Deo N. Prasad
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Distt. Rupnagar, Punjab, 140126, India
| | - Rajesh K. Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Distt. Rupnagar, Punjab, 140126, India
| |
Collapse
|
499
|
Cao SY, Ye SJ, Wang WW, Wang B, Zhang T, Pu YQ. Progress in active compounds effective on ulcerative colitis from Chinese medicines. Chin J Nat Med 2019; 17:81-102. [PMID: 30797423 DOI: 10.1016/s1875-5364(19)30012-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Indexed: 02/09/2023]
Abstract
Ulcerative colitis (UC), a chronic inflammatory disease affecting the colon, has a rising incidence worldwide. The known pathogenesis is multifactorial and involves genetic predisposition, epithelial barrier defects, dysregulated immune responses, and environmental factors. Nowadays, the drugs for UC include 5-aminosalicylic acid, steroids, and immunosuppressants. Long-term use of these drugs, however, may cause several side effects, such as hepatic and renal toxicity, drug resistance and allergic reactions. Moreover, the use of traditional Chinese medicine (TCM) in the treatment of UC shows significantly positive effects, low recurrence rate, few side effects and other obvious advantages. This paper summarizes several kinds of active compounds used in the experimental research of anti-UC effects extracted from TCM, mainly including flavonoids, acids, terpenoids, phenols, alkaloids, quinones, and bile acids from some animal medicines. It is found that the anti-UC activities are mainly focused on targeting inflammation or oxidative stress, which is associated with increasing the levels of anti-inflammatory cytokine (IL-4, IL-10, SOD), suppressing the levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8, IL-23, NF-κB, NO), reducing the activity of MPO, MDA, IFN-γ, and iNOS. This review may offer valuable reference for UC-related studies on the compounds from natural medicines.
Collapse
Affiliation(s)
- Si-Yu Cao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Sheng-Jie Ye
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Wei Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Qiong Pu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
500
|
Liu Y, Zhao J, Zhao Y, Zong S, Tian Y, Chen S, Li M, Liu H, Zhang Q, Jing X, Sun B, Wang H, Sun T, Yang C. Therapeutic effects of lentinan on inflammatory bowel disease and colitis-associated cancer. J Cell Mol Med 2019; 23:750-760. [PMID: 30472806 PMCID: PMC6349230 DOI: 10.1111/jcmm.13897] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/14/2018] [Indexed: 01/03/2023] Open
Abstract
In this study, we investigated the therapeutic potential of lentinan in mouse models of inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). Lentinan decreased the disease activity index and macroscopic and microscopic colon tissue damage in dextran sulphate sodium (DSS)-induced or TNBS-induced models of colitis. High-dose lentinan was more effective than salicylazosulfapyridine in the mouse models of colitis. Lentinan decreased the number of tumours, inflammatory cell infiltration, atypical hyperplasia and nuclear atypia in azoxymethane/DSS-induced CAC model. It also decreased the expression of pro-inflammatory cytokines, such as IL-13 and CD30L, in IBD and CAC model mice possibly by inhibiting Toll-like receptor 4 (TLR4)/NF-κB signalling and the expression of colon cancer markers, such as carcinoembryonic antigen, cytokeratin 8, CK18 and p53, in CAC model mice. In addition, lentinan restored the intestinal bacterial microbiotal community structure in IBD model mice. Thus, it shows therapeutic potential in IBD and CAC model mice possibly by inhibiting TLR4/NF-κB signalling-mediated inflammatory responses and disruption of the intestinal microbiotal structure.
Collapse
Affiliation(s)
- Yanrong Liu
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,Drug Safety Evaluation CenterTianjin International Joint Academy of BiomedicineTianjinChina
| | - Jianmin Zhao
- Department of PathologyHospital of Shun Yi DistrictBeijingChina
| | - Yali Zhao
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Shumin Zong
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Yixuan Tian
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Shuang Chen
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina
| | - Meng Li
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Huijuan Liu
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina
| | - Qiang Zhang
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Xueshuang Jing
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina
| | - Bo Sun
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina
| | - Hongzhi Wang
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Tao Sun
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Cheng Yang
- Tianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjinChina,State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| |
Collapse
|