1
|
He L, Li A, Yu P, Qin S, Tan HY, Zou D, Wu H, Wang S. Therapeutic peptides in the treatment of digestive inflammation: Current advances and future prospects. Pharmacol Res 2024; 209:107461. [PMID: 39423954 DOI: 10.1016/j.phrs.2024.107461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
Digestive inflammation is a widespread global issue that significantly impacts quality of life. Recent advances have highlighted the unique potential of therapeutic peptides for treating this condition, owing to their specific bioactivity and high specificity. By specifically targeting key proteins involved in the pathological process and modulating biomolecular functions, therapeutic peptides offer a novel and promising approach to managing digestive inflammation. This review explores the development history, pharmacological characteristics, clinical applications, and regulatory mechanisms of therapeutic peptides in treating digestive inflammation. Additionally, the review addresses pharmacokinetics and quality control methods of therapeutic peptides, focusing on challenges such as low bioavailability, poor stability, and difficulties in delivery. The role of modern biotechnologies and nanotechnologies in overcoming these challenges is also examined. Finally, future directions for therapeutic peptides and their potential impact on clinical applications are discussed, with emphasis placed on their significant role in advancing medical and therapeutic practices.
Collapse
Affiliation(s)
- Liangliang He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Aijing Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Ping Yu
- Department of Pharmacy, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Shumin Qin
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Hor-Yue Tan
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR
| | - Denglang Zou
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China.
| | - Haomeng Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.
| | - Shuai Wang
- Chinese Medicine Guangdong Laboratory, Hengqin, China; School of Pharmaceutical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
2
|
Siddiqui SH, Khan M, Park J, Lee J, Choe H, Shim K, Kang D. COPA3 peptide supplementation alleviates the heat stress of chicken fibroblasts. Front Vet Sci 2023; 10:985040. [PMID: 36908511 PMCID: PMC9998527 DOI: 10.3389/fvets.2023.985040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/07/2023] [Indexed: 03/14/2023] Open
Abstract
Heat stress inhibits cellular proliferation and differentiation through the production of reactive oxygen species. Under stress conditions, antioxidant drugs promote stable cellular function by reducing the stress level. We sought to demonstrate 9-mer disulfide dimer peptide (COPA3) supplementation stabilizes fibroblast proliferation and differentiation even under heat stress conditions. In our study, fibroblasts were assigned to two different groups based on the temperature, like 38°C group presented as Control - and 43°C group presented as Heat Stress-. Each group was subdivided into two groups depending upon COPA3 treatment, like 38°C + COPA3 group symbolized Control+ and the 43°C + COPA3 group symbolized as Heat Stress+. Heat stress was observed to decrease the fibroblast viability and function and resulted in alterations in the fibroblast shape and cytoskeleton structure. In contrast, COPA3 stabilized the fibroblast viability, shape, and function. Moreover, heat stress and COPA3 were found to have opposite actions with respect to energy production, which facilitates the stabilization of cellular functions by increasing the heat tolerance capacity. The gene expression levels of antioxidant and heat shock proteins were higher after heat stress. Additionally, heat stress promotes the mitogen-activated protein kinase/ extracellular signal-regulated kinase-nuclear factor erythroid 2-related factor 2 (MAPK/ERK-Nrf2). COPA3 maintained the MAPK/ERK-Nrf2 gene expressions that promote stable fibroblast proliferation, and differentiation as well as suppress apoptosis. These findings suggest that COPA3 supplementation increases the heat tolerance capacity, viability, and functional activity of fibroblasts.
Collapse
Affiliation(s)
- Sharif Hasan Siddiqui
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, United States.,Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Mousumee Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Jinryong Park
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Republic of Korea.,Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, Republic of Korea.,3D Tissue Culture Research Center, Konkuk University, Seoul, Republic of Korea
| | - Jeongeun Lee
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Hosung Choe
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Kwanseob Shim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Republic of Korea.,Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Darae Kang
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
3
|
Lee J, Park J, Choe H, Shim K. Insect peptide CopA3 promotes proliferation and PAX7 and MYOD expression in porcine muscle satellite cells. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1132-1143. [PMID: 36812017 PMCID: PMC9890342 DOI: 10.5187/jast.2022.e81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Insects are a valuable natural source that can produce a variety of bioactive compounds due to their increasing species diversity. CopA3 is an antimicrobial peptide derived from Copris tripartitus (i.e., the dung beetle). It is known to increase the proliferation of colonic epithelial and neuronal stem cells by regulating cell cycle. This research hypothesized that CopA3 can promote the proliferation of porcine muscle satellite cells (MSCs). The effects of CopA3 on porcine MSCs, which are important for muscle growth and regeneration, remain unclear. Here, we investigated the effects of CopA3 on porcine MSCs. According to viability results, we designed four groups: control (without CopA3) and three treatment groups (treated with 5,10, and 25 μg/mL of CopA3). At a CopA3 concentration of 5 μg/mL and 10 μg/mL, the proliferation of MSCs increased more than that observed in the control group. Furthermore, compared to that in the control, CopA3 treatment increased the S phase but decreased the G0/G1 phase ratio. Additionally, early and late apoptotic cells were found to be decreased in the 5 μg/mL group. The expressions of the myogenesis-related transcription factor PAX7 and MYOD proteins were significantly upregulated in the 5 μg/mL and 10 μg/mL groups, whereas the MYOG protein remained undetected in all group. This study suggested that CopA3 promotes muscle cell proliferation by regulating the cell cycle of MSCs and can regulate the activity of MSCs by increasing the expressions of PAX7 and MYOD.
Collapse
Affiliation(s)
- Jeongeun Lee
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea
| | - Jinryoung Park
- Department of Stem Cell and Regenerative
Biotechnology, Konkuk University, Seoul 06591, Korea,3D Tissue Culture Research Center, Konkuk
University, Seoul 06591, Korea
| | - Hosung Choe
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| | - Kwanseob Shim
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea,Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea,Corresponding author: Kwanseob Shim,
Department of Agricultural Convergence Technology, Jeonbuk National University,
Jeonju 54896, Korea. Tel: +82-63-270-2609, E-mail:
| |
Collapse
|
4
|
Wu Z, Liu X, Huang S, Li T, Zhang X, Pang J, Zhao J, Chen L, Zhang B, Wang J, Han D. Milk Fat Globule Membrane Attenuates Acute Colitis and Secondary Liver Injury by Improving the Mucus Barrier and Regulating the Gut Microbiota. Front Immunol 2022; 13:865273. [PMID: 35799795 PMCID: PMC9253277 DOI: 10.3389/fimmu.2022.865273] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Inflammatory bowel disease (IBD) often occurs along with extraintestinal manifestations, including hepatic injury. Milk fat globule membrane (MFGM) is an active substance with a potential anti-inflammation activity. However, its alleviated effect and mechanisms in IBD as well as the IBD-induced secondary liver injury are still unclear. Methods C57BL/6J mice were administered with a 21-day oral gavage of MFGM, followed by 7 days of drinking water with 4% dextran sulfate sodium (DSS). Disease activity index (DAI), histological features, and cytokines of the colon and liver were evaluated. Then, RNA-seq of the colon and liver was conducted. The gut microbiota was assessed by analyzing 16S rRNA gene sequences, and finally the integrity and the function of the mucus barrier were evaluated by Alcian blue staining, real-time quantitative PCR, and ELISA. Results Prophylactic MFGM treatment was effective against colitis to include effects in body weight loss, DAI score, colonic length, intestinal pathology, and histological score. Additionally, prophylactic MFGM decreased the levels of interleukin (IL)-1β, IL-6, and myeloperoxidase in colonic tissue, while it increased the IL-10 level. Moreover, the gene expressions of MUC2, MUC4, Reg3b, and Reg3g associated with the production of the molecular mediator of immune response, membrane invagination, and response to protozoan were strikingly upregulated when administered with MFGM. On the other hand, the beneficial effects of MFGM were related to the enriched abundance of genera such as Faccalibacumum and Roseburia in feces samples. Consistently, the administration of MFGM was also found to alleviate DSS-induced hepatic injury. Furthermore, the glutathione transferase activity pathway was enriched in the liver of MFGM-treated mice after DSS administration. Mechanistically, prophylactic MFGM enhanced the mucosal barrier by increasing the gene levels of Reg3b and Reg3g. Meanwhile, the alleviation of MFGM on liver injury was dependent on the reduced hepatic oxidative stress. Conclusions MFGM attenuated colitis and hepatic injury by maintaining the mucosal barrier and bacterial community while inhibiting oxidative stress, which might be an effective therapy of hepatic injury secondary to IBD.
Collapse
Affiliation(s)
- Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiaoyi Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiaman Pang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junying Zhao
- National Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lijun Chen
- National Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Bing Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Dandan Han,
| |
Collapse
|
5
|
Therapeutic peptides: current applications and future directions. Signal Transduct Target Ther 2022; 7:48. [PMID: 35165272 PMCID: PMC8844085 DOI: 10.1038/s41392-022-00904-4] [Citation(s) in RCA: 751] [Impact Index Per Article: 250.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
Peptide drug development has made great progress in the last decade thanks to new production, modification, and analytic technologies. Peptides have been produced and modified using both chemical and biological methods, together with novel design and delivery strategies, which have helped to overcome the inherent drawbacks of peptides and have allowed the continued advancement of this field. A wide variety of natural and modified peptides have been obtained and studied, covering multiple therapeutic areas. This review summarizes the efforts and achievements in peptide drug discovery, production, and modification, and their current applications. We also discuss the value and challenges associated with future developments in therapeutic peptides.
Collapse
|
6
|
Ho LH, Tan TC, Chong LC. Designer foods as an effective approach to enhance disease preventative properties of food through its health functionalities. FUTURE FOODS 2022. [DOI: 10.1016/b978-0-323-91001-9.00031-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
7
|
Dong L, Tomassen MM, Ariëns RMC, Oosterink E, Wichers HJ, Veldkamp T, Mes JJ, Govers C. Clostridioides difficile toxin A-mediated Caco-2 cell barrier damage was attenuated by insect-derived fractions and corresponded to increased gene transcription of cell junctional and proliferation proteins. Food Funct 2021; 12:9248-9260. [PMID: 34606540 DOI: 10.1039/d1fo00673h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pathogenesis of C. difficile in the intestine is associated with the secretion of toxins which can damage the intestinal epithelial layer and result in diseases such as diarrhoea. Treatment for C. difficile infections consists of antibiotics which, however, have non-specific microbiocidal effects and may cause intestinal dysbiosis which results in subsequent health issues. Therefore, alternative treatments to C. difficile infections are required. We investigated whether different black soldier fly- and mealworm-derived fractions, after applying the INFOGEST digestion protocol, could counteract C. difficile toxin A-mediated barrier damage of small intestinal Caco-2 cells. Treatment and pre-treatment with insect-derived fractions significantly (p < 0.05) mitigated the decrease of the transepithelial electrical resistance (TEER) of Caco-2 cells induced by C. difficile toxin A. In relation to these effects, RNA sequencing data showed an increased transcription of cell junctional and proliferation protein genes in Caco-2 cells. Furthermore, the transcription of genes regulating immune signalling was also increased. To identify whether this resulted in immune activation we used a Caco-2/THP-1 co-culture model where the cells were only separated by a permeable membrane. However, the insect-derived fractions did not change the basolateral secreted IL-8 levels in this model. To conclude, our findings suggest that black soldier fly- and mealworm-derived fractions can attenuate C. difficile induced intestinal barrier disruption and they might be promising tools to reduce the symptoms of C. difficile infections.
Collapse
Affiliation(s)
- Liyou Dong
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands. .,Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Monic M Tomassen
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Renata M C Ariëns
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Els Oosterink
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Harry J Wichers
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands. .,Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Teun Veldkamp
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Jurriaan J Mes
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Coen Govers
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands. .,Laboratory of Cell Biology and Immunology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
8
|
Dey DK, Kang SC. CopA3 peptide induces permanent cell-cycle arrest in colorectal cancer cells. Mech Ageing Dev 2021; 196:111497. [PMID: 33957217 DOI: 10.1016/j.mad.2021.111497] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022]
Abstract
Cell-cycle arrest reflects an accumulation of responses to DNA damage that sequentially affects cell growth and division. Herein, we analyzed the effect of the 9-mer dimer defensin-like peptide, CopA3, against colorectal cancer cell growth and proliferation in a dose-dependent manner upon 96 h of treatment. As observed, CopA3 treatment significantly affected cancer cell growth, reduced colony formation ability, increased the number of SA-β-Gal positive cells, and remarkably reduced Ki67 protein expression. Notably, in HCT-116 cells, CopA3 (5 μM) treatment effectively increased oxidative stress and, as a result, amplified the endogenous ROS, mitochondrial ROS, and NO content in the cells, which further activated the DNA damage response and caused cell-cycle arrest at the G1 phase. The prolonged cell-cycle arrest elevated the release of inflammatory cytokines in the cell supernatant. Nevertheless, mechanistically, NAC treatment effectively reversed the CopA3 effect and significantly reduced the oxidative stress; subsequently rescuing the cells from G1 phase arrest. Overall, CopA3 treatment can inhibit the growth and proliferation of colorectal cancer cells by inducing cell-cycle arrest through the ROS-mediated pathway.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
9
|
Kim YH, Hwang JS, Yoon IN, Lee JH, Lee J, Park KC, Seok H, Kim H. The insect peptide CopA3 blocks programmed cell death by directly binding caspases and inhibiting their proteolytic activation. Biochem Biophys Res Commun 2021; 547:82-88. [PMID: 33610044 DOI: 10.1016/j.bbrc.2021.01.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 11/29/2022]
Abstract
Caspases play essential roles in apoptotic processes, which is necessary for cellular homeostasis. However, over-activation of caspases and subsequent excessive apoptosis is considered a main cause of Parkinson's disease and liver diseases. Here, we found that the insect-derived peptide, CopA3, which has shown antiapoptotic effects in many apoptosis models, directly binds to caspases. The resulting complexes do not dissociate during denaturing polyacrylamide gel electrophoresis, as evidenced by a distinct shift in the migration of caspase reflecting an increase in their molecular weight. Surface plasmon resonance and experiment using cysteine-substituted mutants of CopA3 collectively revealed that binding of CopA3 to caspases is dependent on an internal cysteine residue. Notably, CopA3 binding significantly inhibited proteolytic activation of downstream caspases by upstream caspases. In summary, the demonstration that CopA3 directly binds to caspases and inhibits their activating cleavage suggests a possible therapeutic approach for treating human diseases resulting from uncontrolled apoptosis.
Collapse
Affiliation(s)
- Young Ha Kim
- Department of Life Science, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 487-711, Republic of Korea
| | - Jae Sam Hwang
- Department of Agricultural Biology, National Academy of Agricultural Science, RDA, Suwon, 441-707, Republic of Korea
| | - I Na Yoon
- Department of Life Science, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 487-711, Republic of Korea
| | - Joon Ha Lee
- Department of Agricultural Biology, National Academy of Agricultural Science, RDA, Suwon, 441-707, Republic of Korea
| | - Junguee Lee
- Department of Pathology, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-ro 64, Jung-gu, Daejeon, 301-723, Republic of Korea
| | - Ki Cheol Park
- Clinical Research Institute, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Heon Seok
- Korea Brain Bank, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Ho Kim
- Department of Life Science, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 487-711, Republic of Korea.
| |
Collapse
|
10
|
Dey DK, Chang SN, Kang SC. The inflammation response and risk associated with aflatoxin B1 contamination was minimized by insect peptide CopA3 treatment and act towards the beneficial health outcomes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115713. [PMID: 33038573 DOI: 10.1016/j.envpol.2020.115713] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/08/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
This study focused on the possible chemo-preventive effects of insect peptide CopA3 on normal human colon cells against the inflammation induced by the toxic environmental pollutant aflatoxin B1 (AFB1). In the study, we used CCD 841 CoN normal human colon cells to investigate the cytotoxic effect induced by AFB1 and elucidated the negative impact of AFB1 exposure on the cell cycle progression. Further, we also carried out the in-vivo experiment, where male BALB/c mice were administrated with AFB1 to induce inflammation associated cancer like phenotype and the dietary effect of CopA3 was evaluated on the early stages of AFB1-induced hepatotoxicity and inflammation in colon tissues. At the initiation stage, CopA3 was given along with water, which significantly decreased the inflammation in the liver and colon of AFB1 exposed mice model. Mice that received CopA3 alone showed enhanced activity of several antioxidant enzymes. In the post treatment stage, the CopA3 dosage remarkably increased the Ki-67 protein expression, indicating the enhancement in cell proliferation event and increased the number of apoptotic cells in colonic crypts, suggesting the capability of CopA3 treatment towards the epithelial cell turnover. Thus, CopA3 treatment shows its potential to inhibit the development of the early stages of AFB1-induced colon inflammation and hepatotoxicity in mice by inhibiting the DNA synthesis of the damaged and inflammatory cell and induced apoptosis for the clearance of damaged cells. Collectively, the results of this study suggest that CopA3 treatment may play a protective role against the mycotoxin induced inflammation.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Jillyang, Naeri-riGyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sukkum Ngullie Chang
- Department of Biotechnology, Daegu University, Jillyang, Naeri-riGyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Jillyang, Naeri-riGyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
11
|
Wang L, Zhao X, Xia X, Zhu C, Qin W, Xu Y, Hang B, Sun Y, Chen S, Zhang H, Jiang J, Hu J, Fotina H, Zhang G. Antimicrobial Peptide JH-3 Effectively Kills Salmonella enterica Serovar Typhimurium Strain CVCC541 and Reduces Its Pathogenicity in Mice. Probiotics Antimicrob Proteins 2020; 11:1379-1390. [PMID: 31001786 DOI: 10.1007/s12602-019-09533-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Salmonella is an important zoonotic pathogen and is a major cause of gastrointestinal diseases worldwide. The current serious problem of antibiotic abuse has prompted the search for new substitutes for antibiotics. JH-3 is a small antimicrobial peptide with broad-spectrum bactericidal activity. In this study, we showed that JH-3 has good bactericidal activity towards the clinical isolate Salmonella enterica serovar Typhimurium strain CVCC541. The minimum inhibitory concentration (MIC) of JH-3 against this bacterium was determined to be 100 μg/mL, which could decrease the number of CVCC541 cells by 1000-fold in vitro within 5 h. The transmission electron microscopy (TEM) results showed that JH-3 can damage the cell wall and membrane of CVCC541, leading to the leakage of cell contents and subsequent cell death. To measure the bactericidal activity of CVCC541-infected mice were treated intraperitoneally 40 or 10 mg/kg JH-3 at 2 h or 3 days postinfection. Our results showed that treatment with 40 mg/kg JH-3 at 2 h postinfection had the best therapeutic effect and could significantly protect mice from a lethal dose of CVCC541. Furthermore, the clinical symptoms, bacterial burden in blood and organs, and intestinal pathological changes were all decreased and were close to normal. This study examined the therapeutic effect of the antimicrobial peptide JH-3 against S. enterica CVCC541 infection for the first time and determined the therapeutic effect of different JH-3 doses and treatment times, laying the foundation for studies of new antimicrobial agents.
Collapse
Affiliation(s)
- Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China.,College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, People's Republic of China.,Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, People's Republic of China
| | - Xueqin Zhao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China.,Sumy National Agrarian University, Sumy, Ukraine
| | - Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Chunling Zhu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Wanhai Qin
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Yanzhao Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Bolin Hang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Yawei Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Shijun Chen
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Huihui Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Jinqing Jiang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China. .,College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, People's Republic of China.
| | - Hanna Fotina
- Sumy National Agrarian University, Sumy, Ukraine
| | - Gaiping Zhang
- Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, People's Republic of China
| |
Collapse
|
12
|
Liu Y, Perumalsamy H, Kang CH, Kim SH, Hwang JS, Koh SC, Yi TH, Kim YJ. Intracellular synthesis of gold nanoparticles by Gluconacetobacter liquefaciens for delivery of peptide CopA3 and ginsenoside and anti-inflammatory effect on lipopolysaccharide-activated macrophages. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:777-788. [DOI: 10.1080/21691401.2020.1748639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ying Liu
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Chang Ho Kang
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| | - Seung Hyun Kim
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Jae-Sam Hwang
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development, Wanju, Republic of Korea
| | - Sung-Cheol Koh
- Department of Environmental Engineering, Korea Maritime and Ocean University, Busan, Republic of Korea
| | - Tae-Hoo Yi
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| | - Yeon-Ju Kim
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
13
|
Wu K, Li S, Wang J, Ni Y, Huang W, Liu Q, Ling E. Peptide Hormones in the Insect Midgut. Front Physiol 2020; 11:191. [PMID: 32194442 PMCID: PMC7066369 DOI: 10.3389/fphys.2020.00191] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
Insects produce many peptide hormones that play important roles in regulating growth, development, immunity, homeostasis, stress, and other processes to maintain normal life. As part of the digestive system, the insect midgut is also affected by hormones secreted from the prothoracic gland, corpus allatum, and various neuronal cells; these hormones regulate the secretion and activity of insects’ digestive enzymes and change their feeding behaviors. In addition, the insect midgut produces certain hormones when it recognizes various components or pathogenic bacteria in ingested foods; concurrently, the hormones regulate other tissues and organs. In addition, intestinal symbiotic bacteria can produce hormones that influence insect signaling pathways to promote host growth and development; this interaction is the result of long-term evolution. In this review, the types, functions, and mechanisms of hormones working on the insect midgut, as well as hormones produced therein, are reviewed for future reference in biological pest control.
Collapse
Affiliation(s)
- Kai Wu
- College of Life Sciences, Shangrao Normal University, Shangrao, China
| | - Shirong Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- College of Life Sciences, Shangrao Normal University, Shangrao, China
| | - Yuyang Ni
- College of Life Sciences, Shangrao Normal University, Shangrao, China
| | - Wuren Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Qiuning Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.,Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-Agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetland, Yancheng Teachers University, Yancheng, China
| | - Erjun Ling
- Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.,Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Dutta P, Sahu RK, Dey T, Lahkar MD, Manna P, Kalita J. Beneficial role of insect-derived bioactive components against inflammation and its associated complications (colitis and arthritis) and cancer. Chem Biol Interact 2019; 313:108824. [PMID: 31542397 DOI: 10.1016/j.cbi.2019.108824] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/19/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
Insect-based bioactive components are emerging as novel sources of drugs, effective against various diseases. Inflammation is considered to be an innate immune response developed by different organisms against foreign pathogens and cellular stress. However, repetitive elevated inflammation is considered to be responsible for development of many other diseases including colitis and arthritis. Due to the limited activities and side effects of non-steroidal anti-inflammatory drugs, researchers are continuously looking for alternative sources of drug molecules to alleviate the inflammatory related complications. Recently, insect-based bioactive components, such as venoms, haemocytes, cecropin A, papiliocin, N-acetyldopamine dimers, cecropin-TY1 peptide, cop A3 peptide, glycosaminoglycan, coprisin peptide, silk fibroin microparticles, and silk fibroin nanoparticles have been found to be active against different inflammatory mechanisms and associated diseases. Cancers, are some of the deadliest diseases, which are mainly treated by chemotherapy, radiation therapy and surgery. However, such treatments, mainly chemotherapy, is associated with enormous side effects. Therefore, as an alternative, less hazardous option, compounds from insects with anti-cancerous activity are being explored. Insect-derived compounds, such as cantharidin, norcantharidin, isocoumarin, plancyols A, plancypyrazine A, pancratistatin, narciclasine, and ungeremine, show potential anti-cancerous activity. In this review, we will be discussing the role of different potential drug molecules of insect origin with special emphasis on anti-inflammation and their association with health disorders and cancer.
Collapse
Affiliation(s)
- Prachurjya Dutta
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| | - Ravi Kumar Sahu
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Tapan Dey
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India; Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Manisha Datta Lahkar
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India
| | - Prasenjit Manna
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Jatin Kalita
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology (CSIR-NEIST), Jorhat, 785006, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
15
|
Chen T, Huang H, Zhou Y, Geng L, Shen T, Yin S, Zhou L, Zheng S. HJURP promotes hepatocellular carcinoma proliferation by destabilizing p21 via the MAPK/ERK1/2 and AKT/GSK3β signaling pathways. J Exp Clin Cancer Res 2018; 37:193. [PMID: 30111352 PMCID: PMC6094877 DOI: 10.1186/s13046-018-0866-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/06/2018] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Holliday junction recognition protein (HJURP) has been implicated in many cancers including hepatocellular carcinoma (HCC). However, the underlying mechanism by which HJURP promotes HCC cell proliferation remains unclear. METHODS RT-qPCR and immunohistochemistry were used to detect HJURP expression in HCC and adjacent tumor tissues and HCC cell lines. The localization of p21 were determined by immunofluorescence and western blot. Co-immunoprecipitation and western blot were used to validate the p21 stability and signaling pathways affected by HJURP. The effects of HJURP on HCC cell proliferation were assessed both in vivo and in vitro. The ERK1/2 pathway inhibitor U0126 and AKT pathway agonist SC-79 were used to treat HCC cell lines for further mechanistic investigations. RESULTS HJURP expression was higher in HCC tissues than in para-tumor tissues. Moreover, ectopic HJURP expression facilitated the proliferation of HCC cells, whereas the depletion of HJURP resulted in decreased cell growth in vitro and in vivo. Furthermore, the effects of HJURP silencing were reversed by p21 knockdown. Likewise, p21 overexpression inhibited cell growth ability mediated by HJURP elevation. Mechanistically, HJURP destabilized p21 via the MAPK/ERK1/2 and AKT/GSK3β pathways, which regulated the nucleus-cytoplasm translocation and ubiquitin-mediated degradation of p21. Clinically, high HJURP expression was correlated with unfavorable prognoses in HCC individuals. CONCLUSIONS Our data revealed that HJURP is an oncogene that drives cell cycle progression upstream of p21 in HCC. These findings may provide a potential therapeutic and prognostic target for HCC.
Collapse
Affiliation(s)
- Tianchi Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Hechen Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Yuan Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Lei Geng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tian Shen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengyong Yin
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
| | - Lin Zhou
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, Hangzhou, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Yoon IN, Lu LF, Hong J, Zhang P, Kim DH, Kang JK, Hwang JS, Kim H. The American cockroach peptide periplanetasin-4 inhibits Clostridium difficile toxin A-induced cell toxicities and inflammatory responses in the mouse gut. J Pept Sci 2017; 23:833-839. [PMID: 28949065 DOI: 10.1002/psc.3046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/27/2017] [Accepted: 08/27/2017] [Indexed: 01/07/2023]
Abstract
Many reports have shown that crude extracts of the American cockroach have therapeutic effects on inflammation. In a previous study, our research group showed that an antimicrobial peptide (Periplanetasin-2) derived from the American cockroach via de novo transcriptome analysis inhibited apoptosis of human colonocytes and inflammatory responses of the mouse gut caused by Clostridium difficile toxin A. Here, we examined whether Periplanetasin-4 (Peri-4), another antimicrobial peptide identified via de novo transcriptome analysis of the American cockroach, could also inhibit the various toxicities induced by C. difficile toxin A. We found that Peri-4 significantly reduced the cell viability loss and cell apoptosis caused by toxin A in vitro. Peri-4 also ameliorated the severe inflammatory responses seen in the toxin A-induced mouse enteritis model, rescuing the villus disruption and interleukin-6 production induced by luminal injection of toxin A into the mouse gut. Mechanistically, we found that Peri-4 could reduce toxin A-induced reactive oxygen species production to inhibit the activations of p38MAPK and p21Cip1/Waf1 , which are critical for the cell damages induced by toxin A. These results collectively suggest that the Peri-4 may be a potential therapeutic agent for treating toxin A-induced pseudomembranous colitis. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- I Na Yoon
- Division of Life Science and Chemistry, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 11159, Korea
| | - Li Fang Lu
- Hainan Institute of Science and Technology, Haikou, 571126, China
| | - Ji Hong
- Division of Life Science and Chemistry, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 11159, Korea
| | - Peng Zhang
- Division of Life Science and Chemistry, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 11159, Korea
| | - Dae Hong Kim
- Division of Life Science and Chemistry, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 11159, Korea
| | - Jin Ku Kang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University Graduate School of Medicine, Incheon, 406-840, Korea
| | - Jae Sam Hwang
- Department of Agricultural Biology, National Academy of Agricultural Science, RDA, Wanju, 55365, Korea
| | - Ho Kim
- Division of Life Science and Chemistry, College of Natural Science, Daejin University, Pocheon, Gyeonggido, 11159, Korea
| |
Collapse
|