1
|
Bullard BM, McDonald SJ, Cardaci TD, VanderVeen BN, Mohammed AD, Kubinak JL, Pierre JF, Chatzistamou I, Fan D, Hofseth LJ, Murphy EA. Panaxynol improves crypt and mucosal architecture, suppresses colitis-enriched microbes, and alters the immune response to mitigate colitis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G591-G606. [PMID: 38469632 PMCID: PMC11376977 DOI: 10.1152/ajpgi.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory disease of the large intestine, which impacts millions worldwide. Current interventions aimed at treating UC symptoms can have off-target effects, invoking the need for alternatives that may provide similar benefits with less unintended consequences. This study builds on our initial data, which showed that panaxynol-a novel, potent, bioavailable compound found in American ginseng-can suppress disease severity in murine colitis. Here we explore the underlying mechanisms by which panaxynol improves both chronic and acute murine colitis. Fourteen-week-old C57BL/6 female mice were either given three rounds of dextran sulfate sodium (DSS) in drinking water to induce chronic colitis or one round to induce acute colitis. Vehicle or panaxynol (2.5 mg/kg) was administered via oral gavage three times per week for the study duration. Consistent with our previous findings, panaxynol significantly (P < 0.05) improved the disease activity index and endoscopic scores in both models. Using the acute model to examine potential mechanisms, we show that panaxynol significantly (P < 0.05) reduced DSS-induced crypt distortion, goblet cell loss, and mucus loss in the colon. 16S Sequencing revealed panaxynol altered microbial composition to suppress colitis-enriched genera (i.e., Enterococcus, Eubacterium, and Ruminococcus). In addition, panaxynol significantly (P < 0.05) suppressed macrophages and induced regulatory T-cells in the colonic lamina propria. The beneficial effects of panaxynol on mucosal and crypt architecture, combined with its microbial and immune-mediated effects, provide insight into the mechanisms by which panaxynol suppresses murine colitis. Overall, this data is promising for the use of panaxynol to improve colitis in the clinic.NEW & NOTEWORTHY In the current study, we report that panaxynol ameliorates chemically induced murine colitis by improving colonic crypt and mucosal architecture, suppressing colitis-enriched microbes, reducing macrophages, and promoting the differentiation of regulatory T-cells in the colonic lamina propria. This study suggests that this novel natural compound may serve as a safe and effective treatment option for colitis patients.
Collapse
Affiliation(s)
- Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Sierra J McDonald
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Thomas D Cardaci
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Ahmed D Mohammed
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Jason L Kubinak
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| |
Collapse
|
2
|
Zhao L, Zhang T, Zhang K. Pharmacological effects of ginseng and ginsenosides on intestinal inflammation and the immune system. Front Immunol 2024; 15:1353614. [PMID: 38698858 PMCID: PMC11064651 DOI: 10.3389/fimmu.2024.1353614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Intestinal inflammatory imbalance and immune dysfunction may lead to a spectrum of intestinal diseases, such as inflammatory bowel disease (IBD) and gastrointestinal tumors. As the king of herbs, ginseng has exerted a wide range of pharmacological effects in various diseases. Especially, it has been shown that ginseng and ginsenosides have strong immunomodulatory and anti-inflammatory abilities in intestinal system. In this review, we summarized how ginseng and various extracts influence intestinal inflammation and immune function, including regulating the immune balance, modulating the expression of inflammatory mediators and cytokines, promoting intestinal mucosal wound healing, preventing colitis-associated colorectal cancer, recovering gut microbiota and metabolism imbalance, alleviating antibiotic-induced diarrhea, and relieving the symptoms of irritable bowel syndrome. In addition, the specific experimental methods and key control mechanisms are also briefly described.
Collapse
Affiliation(s)
| | | | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
McDonald SJ, Bullard BM, VanderVeen BN, Cardaci TD, Huss AR, Fan D, Hofseth LJ, Murphy EA. Panaxynol alleviates colorectal cancer in a murine model via suppressing macrophages and inflammation. Am J Physiol Gastrointest Liver Physiol 2023; 325:G318-G333. [PMID: 37489869 PMCID: PMC10642997 DOI: 10.1152/ajpgi.00119.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023]
Abstract
Currently available colorectal cancer (CRC) therapies have limited efficacy and severe adverse effects that may be overcome with the alternative use of natural compounds. We previously reported that panaxynol (PA), a bioactive component in American ginseng, possesses anticancer properties in vitro and suppresses murine colitis through its proapoptotic and anti-inflammatory properties. Because colitis is a predisposing factor of CRC and inflammation is a major driver of CRC, we sought to evaluate the therapeutic potential of PA in CRC. Azoxymethane-dextran sodium sulfate (AOM/DSS) mice (C57BL/6) were administered 2.5 mg/kg PA or vehicle 3 times/wk via oral gavage over 12 wk. PA improved clinical symptoms (P ≤ 0.05) and reduced tumorigenesis (P ≤ 0.05). This improvement may be reflective of PA's restorative effect on intestinal barrier function; PA upregulated the expression of essential tight junction and mucin genes (P ≤ 0.05) and increased the abundance of mucin-producing goblet cells (P ≤ 0.05). Given that macrophages play a substantial role in the pathogenesis of CRC and that we previously demonstrated that PA targets macrophages in colitis, we next assessed macrophages. We show that PA reduces the relative abundance of colonic macrophages within the lamina propria (P ≤ 0.05), and this was consistent with a reduction in the expression of important markers of macrophages and inflammation (P ≤ 0.05). We further confirmed PA's inhibitory effects on macrophages in vitro under CRC conditions (P ≤ 0.05). These results suggest that PA is a promising therapeutic compound to treat CRC and improve clinical symptoms given its ability to inhibit macrophages and modulate the inflammatory environment in the colon.NEW & NOTEWORTHY We report that panaxynol (PA) reduces colorectal cancer (CRC) by improving the colonic and tumor environment. Specifically, we demonstrate that PA improves crypt morphology, upregulates crucial tight junction and mucin genes, and promotes the abundance of mucin-producing goblet cells. Furthermore, PA reduces macrophages and associated inflammation, important drivers of CRC, in the colonic environment. This present study provides novel insights into the potential of PA as a therapeutic agent to ameliorate CRC tumorigenesis.
Collapse
Affiliation(s)
- Sierra J McDonald
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Brooke M Bullard
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Thomas D Cardaci
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Alexander R Huss
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, South Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| |
Collapse
|
4
|
Pu J, Zhou X, Liu J, Hou P, Ji M. Therapeutic potential and deleterious effect of glucocorticoids on azoxymethane/dextran sulfate sodium-induced colorectal cancer in mice. Am J Cancer Res 2021; 11:4866-4883. [PMID: 34765297 PMCID: PMC8569368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023] Open
Abstract
Glucocorticoids (GCs) are widely used in the treatment of various autoimmune and inflammatory diseases, including inflammatory bowel disease (IBD). However, the effect of GCs on the progression of colitis-associated colorectal cancer (CAC) has not been well explored. In this study, we first established a colorectal cancer model induced by azoxymethane and dextran sulfate sodium (AOM/DSS) and a colitis model induced by DSS in mice. Dexamethasone (DEX) was then administered at different periods of time to determine its effect on tumorigenesis and tumor progression. Meanwhile, body weight, stool property and fecal blood of mice were recorded. At the end of this study, the number and load of tumors were evaluated, and the expression of proteins associated with cell proliferation was analyzed. To evaluate the inflammation in colon, we detected the level of pro-inflammatory cytokine TNFα, and the mucosal infiltration of inflammatory cells. Our results revealed that AOM injection followed by three cycles of drinking water containing 1.5% DSS successfully induced multiple tumor formation in mouse colon and rectum. Both early and late DEX intervention suppressed tumor growth in mouse colorectum, and downregulated the expression of PCNA and cyclin D1. Moreover, DEX treatment significantly inhibited TNFα production, mucosal infiltration of inflammatory cells, and the activity of MAPK/JNK pathway, particularly early DEX intervention. However, we also found that DEX treatment deteriorated the general state of mouse manifested by greater loss of body weight and rectal bleeding. In summary, both early and late DEX interventions significantly ameliorate colonic inflammation and inhibit the progression of AOM/DSS-induced colorectal cancer, at least partly due to the inhibition of MAPK/JNK pathway. It is noteworthy that the deleterious effect on the general condition of mouse may limit the duration of GCs treatment.
Collapse
Affiliation(s)
- Jun Pu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Xinrui Zhou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Jiaxin Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Peng Hou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| |
Collapse
|
5
|
Zhao H, He M, Zhang M, Sun Q, Zeng S, Chen L, Yang H, Liu M, Ren S, Meng X, Xu H. Colorectal Cancer, Gut Microbiota and Traditional Chinese Medicine: A Systematic Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:805-828. [PMID: 33827382 DOI: 10.1142/s0192415x21500385] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Based on the study and research on the pathogenesis of colorectal cancer, the types and functions of gut microbiota, and its role in guiding and regulating the occurrence and development of diseases, we have explored the mechanism of traditional Chinese medicine in the treatment of colorectal cancer by regulating the gut microbiota. Genetic variation, abnormal responses of innate and adaptive immunity, mucosal barrier dysfunction, imbalance of intestinal microbial colonization, personal and environmental risk factors are the main pathogenesis of colorectal cancer. The gut microbiota mainly includes Sclerotium (including Clostridium, Enterococcus, Lactobacillus and Ruminococcus) and Bacteroides (including Bacteroides and Prevotella), which have biological antagonism, nutrition for the organism, metabolic abilities, immune stimulation, and ability to shape cancer genes functions to body. The gut microbiota can be related to the health of the host. Current studies have shown that Chinese herbal compound, single medicinal materials, and monomer components can treat colorectal cancer by regulating the gut microbiota, such as Xiaoyaosan can increase the abundance of Bacteroides, Lactobacillus, and Proteus and decrease the abundance of Desulfovibrio and Rickerella. Therefore, studying the regulation and mechanism of gut microbiota on colorectal cancer is of great benefit to disease treatment.
Collapse
Affiliation(s)
- Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Man He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Meng Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sha Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
6
|
Wada Y, Tokuda K, Morine Y, Okikawa S, Yamashita S, Ikemoto T, Imura S, Saito Y, Yamada S, Shimada M. The inhibitory effect of TU-100 on hepatic stellate cell activation in the tumor microenvironment. Oncotarget 2020; 11:4593-4604. [PMID: 33346211 PMCID: PMC7733620 DOI: 10.18632/oncotarget.27835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION The tumor microenvironment is involved in acquiring tumor malignancies of colorectal liver metastasis (CRLM). We have reported that TU-100 (Daikenchuto) suppresses hepatic stellate cell (HSC) activation in obstructive jaundice. In this study, we report new findings as the direct and indirect inhibitory effects of TU-100 on cancer cell growth through the suppression of HSC activation. MATERIALS AND METHODS The HSCs (LX2) were cultured in colon cancer cells (HCT116 and HT29)-conditioned medium (CM) with or without TU-100 treatment (90, 270, 900 μg/ml). Activated HSCs (aHSCs) were detected by α-SMA and IL-6 mRNA expressions and cytokine arrays of HSC's culture supernatants. Cancer cell growth was analyzed for proliferation and migration ability, compared with TU-100 treatment. To investigate the direct anti-tumor effect of TU-100, cancer cells were cultured in the presence of aHSC-CM and TU-100 (90, 270, 900) or aHSC-CM alone, and assessed autophagosomes, conversion to LC3-II protein, and Beclin-1 mRNA expression. RESULTS Colon cancer-CM significantly increased α-SMA and IL-6 mRNA expressions of aHSC. α-SMA and IL-6 mRNA expressions of aHSC, and IL-6 secretions from aHSCs were significantly decreased with TU-100 (270, 900) treatment, compared to colon cancer-CM alone. Compared with normal culture medium, aHSC-CM led to a significantly increased cell number and modified HSC-CM (TU-100; 270, 900) significantly suppressed cancer cell growth and migration. TU-100 (900) treatment induced autophagy and significantly promoted the autophagic cell death. CONCLUSIONS TU-100 inhibited colon cancer cell malignant potential by both suppressing HSC activation and inducing directly autophagy of cancer cells.
Collapse
Affiliation(s)
- Yuma Wada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,These authors contributed equally to this work
| | - Kazunori Tokuda
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,These authors contributed equally to this work
| | - Yuji Morine
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shohei Okikawa
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shoko Yamashita
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Satoru Imura
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yu Saito
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shinichiro Yamada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Surgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
7
|
Molecules from American Ginseng Suppress Colitis through Nuclear Factor Erythroid-2-Related Factor 2. Nutrients 2020; 12:nu12061850. [PMID: 32575883 PMCID: PMC7353434 DOI: 10.3390/nu12061850] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 01/25/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease that affects millions of people worldwide and increases the risk of colorectal cancer (CRC) development. We have previously shown that American ginseng (AG) can treat colitis and prevent colon cancer in mice. We further fractionated AG and identified the most potent fraction, hexane fraction (HAG), and the most potent compound in this fraction, panaxynol (PA). Because (1) oxidative stress plays a significant role in the pathogenesis of colitis and associated CRC and (2) nuclear factor erythroid-2-related factor 2 (Nrf2) is the master regulator of antioxidant responses, we examined the role of Nrf2 as a mechanism by which AG suppresses colitis. Through a series of in vitro and in vivo Nrf2 knockout mouse experiments, we found that AG and its components activate the Nrf2 pathway and decrease the oxidative stress in macrophages (mΦ) and colon epithelial cells in vitro. Consistent with these in vitro results, the Nrf2 pathway is activated by AG and its components in vivo, and Nrf2-/- mice are resistant to the suppressive effects of AG, HAG and PA on colitis. Results from this study establish Nrf2 as a mediator of AG and its components in the treatment of colitis.
Collapse
|
8
|
Tashkandi H, Chaparala A, Peng S, Nagarkatti M, Nagarkatti P, Chumanevich AA, Hofseth LJ. Pharmacokinetics of Panaxynol in Mice. ACTA ACUST UNITED AC 2020; 4:133-143. [PMID: 32905447 PMCID: PMC7472592 DOI: 10.26502/jcsct.5079059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The purpose of our study is to explore the pharmacokinetic parameters of panaxynol (PA) and understand its potential and dosage used in pre-clinical animal models. For in vitro analysis,5 μM of PA was added to liver microsomes of mouse and human species. Nicotinamide adenine dinucleotide phosphate was added to initiate enzyme reaction except for the negative control. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to measure concentrations. For in vivo studies, CD-1 mice were treated with PA by intravenous (IV) injection or oral administration (PO). Concentrations of PA were measured in plasma and tissue using LC-MS/MS. Pharmacokinetic parameters were obtained using non-compartmental analysis. Area under the curve concentration versus time was calculated using a linear trapezoidal model.In vitro, PA's half-life is 21.4 min and 48.1 min in mouse and human liver microsomes, respectively. In vivo, PA has a half-life of 1.5 hr when IV-injected, and 5.9 hr when administered via PO, with a moderate bioavailability of 50.4%. Mice show no signs of toxicity up to 300 mg/kg PO. PA concentrations were highest in colon tissue 2 hr post-treatment at 486 ng/g of colon tissue.PA's pharmacokinetic properties and low toxicity point to the safety and compatibility of PA with mice.
Collapse
Affiliation(s)
- Hossam Tashkandi
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
- Corresponding Author: Mr. Hossam Tashkandi, Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA, Tel: +1 (803) 381-7220; (or)
| | - Anusha Chaparala
- OB/GYN, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sean Peng
- Touchstone Biosciences, Plymouth Meeting, PA, USA
| | - Mitzi Nagarkatti
- Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Prakash Nagarkatti
- Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Alexander A. Chumanevich
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Lorne J. Hofseth
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
9
|
Oh G, Cho HJ, Suh S, Lee D, Kim K. Multicolor fluorescence imaging using a single RGB-IR CMOS sensor for cancer detection with smURFP-labeled probiotics. BIOMEDICAL OPTICS EXPRESS 2020; 11:2951-2963. [PMID: 32637234 PMCID: PMC7316003 DOI: 10.1364/boe.391417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/16/2020] [Accepted: 04/26/2020] [Indexed: 05/17/2023]
Abstract
A multicolor fluorescence imaging device was recently developed for image-guided surgery. However, conventional systems are typically bulky and function with two cameras. To overcome these issues, we developed an economical home-built fluorescence imaging device based on a single RGB-IR sensor that can acquire both color and fluorescence images simultaneously. The technical feasibility of RGB-IR imaging was verified ex vivo in chicken breast tissue using fluorescein isothiocyanate (FITC), cyanine 5 (Cy5), and indocyanine green (ICG) as fluorescent agents. The minimum sensitivities for FITC, Cy5, and ICG were 0.200 µM, 0.130 µM, and 0.065 µM, respectively. In addition, we validated the fluorescence imaging of this device in vitro during a minimally invasive procedure using smURFP-labeled probiotics, which emit a spectrum similar to that of Cy5. Our preliminary study of the ex vivo tissue suggests that Cy5 and ICG are good candidates for deep tissue imaging. In addition, the tumor-specific amplification process was visualized using cancer cells incubated with probiotics that had been labeled with a fluorescent protein. Our approach indicates the potential for in vivo screening of tumors in rodent tumor models.
Collapse
Affiliation(s)
- Gyungseok Oh
- Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
| | - Hong Jun Cho
- Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
- School of Mechanical Engineering, Korea University and Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
| | - SeungBeum Suh
- Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
| | - Deukhee Lee
- Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
- University of Science and Technology (UST), South Korea the Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, South Korea
| | - Keri Kim
- Center for Medical Robotics, Korea Institute of Science and Technology, South Korea
- University of Science and Technology (UST), South Korea the Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
10
|
Li B, Wang Y, Yin L, Huang G, Xu Y, Su J, Ma L, Lu J. Glucocorticoids promote the development of azoxymethane and dextran sulfate sodium-induced colorectal carcinoma in mice. BMC Cancer 2019; 19:94. [PMID: 30665389 PMCID: PMC6341596 DOI: 10.1186/s12885-019-5299-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 01/09/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Stress has been suggested as a promoter of tumor growth and development. Glucocorticoids (GCs) are the main stress hormones and widely prescribed as drugs. However, the effect of GCs on the development and progression of colorectal carcinoma (CRC) is unclear. METHODS We evaluated the effect of corticosterone (CORT) on azoxymethane and dextran sulfate sodium (AOM/DSS)-induced carcinogenesis in the colorectum of C57BL/6 strain mice. Plasma level of CORT was detected by radioimmunoassay. The expression of proliferation markers (Ki-67 and PCNA), nuclear factor (NF)-κB p65 and phosphoto-p65 (P-p65), as well as cyclooxygenase (COX)-2 were determined by immunohistochemistry. Inflammation in colorectum was evaluated by histopathology. RESULTS CORT feeding in drinking water of mice not only significantly elevated plasma CORT concentration, but also significantly increased the incidence and neoplasms burden (number and size of neoplasms) in colorectum. CORT also significant enhanced the expression of cell proliferation marker (Ki-67 and PCNA), NF-κB p65 and P-p65 as well as COX-2 in colorectal neoplasm of AOM/DSS-treated mice. CONCLUSION In this study, we have found for the first time that CORT at stress level potentially promotes the growth and development of AOM/DSS-induced colorectal adenoma and carcinoma in mice. Up-regulation of NF-κB and COX-2 may be involved in the promoting effect of CORT.
Collapse
Affiliation(s)
- Bo Li
- Department of pathophysiology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China.,Department of general surgery, Changhai hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Yan Wang
- Department of pathophysiology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Lijuan Yin
- Department of pathology, Changhai hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Gaoxiang Huang
- Department of pathophysiology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Yi Xu
- Department of pathology, Changhai hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Jie Su
- Department of pathophysiology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China
| | - Liye Ma
- Department of general surgery, Changhai hospital, Second Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Jian Lu
- Department of pathophysiology, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
11
|
Looking for the best anti-colitis medicine: A comparative analysis of current and prospective compounds. Oncotarget 2018; 8:228-237. [PMID: 27974688 PMCID: PMC5352114 DOI: 10.18632/oncotarget.13894] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/15/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic lifelong inflammatory disorder of the colon, which, while untreated, has a relapsing and remitting course with increasing risk of progression toward colorectal cancer. Current medical treatment strategies of UC mostly focus on inhibition of the signs and symptoms of UC to induce remission and prevent relapse of disease activity, minimizing the impact on quality of life, but not affecting the cause of disease. To date, however, there is no single reliable treatment agent and/or strategy capable of effectively controlling colitis progression throughout the patient's life without side effects, remission, or resistance. Taking into consideration an urgent need for the new colitis treatment strategies, targets and/or modulators of inflammation, we have tested current and prospective compounds for colitis treatment and directly compared their anti-colitis potency using a dextran sulfate sodium (DSS) mouse model of colitis. We have introduced a composite score - a multi-parameters comparison tool - to assess biological potency of different compounds.
Collapse
|
12
|
Li KF, Kang CM, Yin XF, Li HX, Chen ZY, Li Y, Zhang Q, Qiu YR. Ginsenoside Rh2 inhibits human A172 glioma cell proliferation and induces cell cycle arrest status via modulating Akt signaling pathway. Mol Med Rep 2017; 17:3062-3068. [PMID: 29207171 PMCID: PMC5783527 DOI: 10.3892/mmr.2017.8193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023] Open
Abstract
Ginsenoside Rh2 (G-Rh2), the main bioactive component in American ginseng, is known to exert a wide variety of biological activities. Accumulating evidence suggests that G-Rh2 inhibits cell proliferation and induces apoptosis of tumor cells. However, the possible mechanism through which G-Rh2 exerts its action on malignant glioma cells have not been completely elucidated. The findings of the present study demonstrated that G-Rh2 decreased the viability of glioma cells in a dose- and time-dependent manner, and induced cell cycle arrest. G-Rh2-induced cell cycle arrest was accompanied by the downregulation of cyclin-dependent kinase 4 and Cyclin E. In addition, G-Rh2 markedly reduced the expression of total- RAC-α serine/threonine-protein kinase (Akt) and the levels of phosphorylated-Akt. These findings provide mechanistic details of how G-Rh2 acts on glioma cells and suggest that G-Rh2 may function as a potential anti-cancer drug for glioma treatment.
Collapse
Affiliation(s)
- Kai-Fei Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chun-Min Kang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiao-Feng Yin
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hai-Xia Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhuo-Yu Chen
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yu-Rong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
13
|
Hasebe T, Matsukawa J, Ringus D, Miyoshi J, Hart J, Kaneko A, Yamamoto M, Kono T, Fujiya M, Kohgo Y, Wang CZ, Yuan CS, Bissonnette M, Musch MW, Chang EB. Daikenchuto (TU-100) Suppresses Tumor Development in the Azoxymethane and APC min/+ Mouse Models of Experimental Colon Cancer. Phytother Res 2017; 31:90-99. [PMID: 27730672 PMCID: PMC5590753 DOI: 10.1002/ptr.5735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/26/2016] [Accepted: 09/18/2016] [Indexed: 02/06/2023]
Abstract
Chemopreventative properties of traditional medicines and underlying mechanisms of action are incompletely investigated. This study demonstrates that dietary daikenchuto (TU-100), comprised of ginger, ginseng, and Japanese pepper effectively suppresses intestinal tumor development and progression in the azoxymethane (AOM) and APCmin/+ mouse models. For the AOM model, TU-100 was provided after the first of six biweekly AOM injections. Mice were sacrificed at 30 weeks. APCmin/+ mice were fed diet without or with TU-100 starting at 6 weeks, and sacrificed at 24 weeks. In both models, dietary TU-100 decreased tumor size. In APC min/+ mice, the number of small intestinal tumors was significantly decreased. In the AOM model, both TU-100 and Japanese ginseng decreased colon tumor numbers. Decreased Ki-67 and β-catenin immunostaining and activation of numerous transduction pathways involved in tumor initiation and progression were observed. EGF receptor expression and stimulation/phosphorylation in vitro were investigated in C2BBe1 cells. TU-100, ginger, and 6-gingerol suppressed EGF receptor induced Akt activation. TU-100 and ginseng and to a lesser extent ginger or 6-gingerol inhibited EGF ERK1/2 activation. TU-100 and some of its components and metabolites of these components inhibit tumor progression in two mouse models of colon cancer by blocking downstream pathways of EGF receptor activation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Takumu Hasebe
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Jun Matsukawa
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Daina Ringus
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Jun Miyoshi
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Atsushi Kaneko
- Tsumura Research Laboratories, Tsumura and Co., Ami, Ibaraki, Japan
| | | | - Toru Kono
- Center for Clinical and Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Hokkaido, Japan
- Division of Gastroenterologic and General Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Chong-Zi Wang
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Mark W. Musch
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Eugene B. Chang
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Yang J, Yuan D, Xing T, Su H, Zhang S, Wen J, Bai Q, Dang D. Ginsenoside Rh2 inhibiting HCT116 colon cancer cell proliferation through blocking PDZ-binding kinase/T-LAK cell-originated protein kinase. J Ginseng Res 2016; 40:400-408. [PMID: 27746693 PMCID: PMC5052442 DOI: 10.1016/j.jgr.2016.03.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/17/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ginsenoside Rh2 (GRh2) is the main bioactive component in American ginseng, a commonly used herb, and its antitumor activity had been studied in previous studies. PDZ-binding kinase/T-LAK cell-originated protein kinase (PBK/TOPK), a serine/threonine protein kinase, is highly expressed in HCT116 colorectal cancer cells. METHODS We examined the effect of GRh2 on HCT116 cells ex vivo. Next, we performed in vitro binding assay and in vitro kinase assay to search for the target of GRh2. Furthermore, we elucidated the underlying molecular mechanisms for the antitumor effect of GRh2 ex vivo and in vivo. RESULTS The results of our in vitro studies indicated that GRh2 can directly bind with PBK/TOPK and GRh2 also can directly inhibit PBK/TOPK activity. Ex vivo studies showed that GRh2 significantly induced cell death in HCT116 colorectal cancer cells. Further mechanistic study demonstrated that these compounds inhibited the phosphorylation levels of the extracellular regulated protein kinases 1/2 (ERK1/2) and (H3) in HCT116 colorectal cancer cells. In vivo studies showed GRh2 inhibited the growth of xenograft tumors of HCT116 cells and inhibited the phosphorylation levels of the extracellular regulated protein kinases 1/2 and histone H3. CONCLUSION The results indicate that GRh2 exerts promising antitumor effect that is specific to human HCT116 colorectal cancer cells through inhibiting the activity of PBK/TOPK.
Collapse
Affiliation(s)
- Jianjun Yang
- Department of interventional radiology, Affiliated Hospital of Yan'an University, Shanxi, China
| | - Donghong Yuan
- Department of interventional radiology, Affiliated Hospital of Yan'an University, Shanxi, China
| | - Tongchao Xing
- Department of General Surgery, The Fourth People's Hospital, Shanxi, China
| | - Hongli Su
- Department of Anesthesiology, Yanan University Affiliated Hospital, Shanxi, China
| | - Shengjun Zhang
- Department of General Surgery, Yanan University Affiliated Hospital, Shanxi, China
| | - Jiansheng Wen
- Department of Radiology, Central Hospital of Tongchuan Mining Bureau, Shanxi, China
| | - Qiqiang Bai
- Department of Pharmacy, Yanan University Affiliated Hospital, Shanxi, China
| | - Dongmei Dang
- Department of Pathogenic microbiology, Medical College of Yan'an University, Shanxi, China
| |
Collapse
|
15
|
Pabla B, Bissonnette M, Konda VJ. Colon cancer and the epidermal growth factor receptor: Current treatment paradigms, the importance of diet, and the role of chemoprevention. World J Clin Oncol 2015; 6:133-141. [PMID: 26468449 PMCID: PMC4600187 DOI: 10.5306/wjco.v6.i5.133] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/23/2015] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer represents the third most common and the second deadliest type of cancer for both men and women in the United States claiming over 50000 lives in 2014. The 5-year survival rate for patients diagnosed with metastatic colon and rectal cancer is < 15%. Early detection and more effective treatments are urgently needed to reduce morbidity and mortality of patients afflicted with this disease. Here we will review the risk factors and current treatment paradigms for colorectal cancer, with an emphasis on the role of chemoprevention as they relate to epidermal growth factor receptor (EGFR) blockade. We will discuss how various EGFR ligands are upregulated in the presence of Western diets high in saturated and N-6 polyunsaturated fats. We will also outline the various mechanisms of EGFR inhibition that are induced by naturally occurring chemopreventative agents such as ginseng, green tea, and curcumin. Finally, we will discuss the current role of targeted chemotherapy in colon cancer and outline the limitations of our current treatment options, describing mechanisms of resistance and escape.
Collapse
|
16
|
Xie G, Wang CZ, Yu C, Qiu Y, Wen XD, Zhang CF, Yuan CS, Jia W. Metabonomic Profiling Reveals Cancer Chemopreventive Effects of American Ginseng on Colon Carcinogenesis in Apc(Min/+) Mice. J Proteome Res 2015; 14:3336-47. [PMID: 26136108 PMCID: PMC6098237 DOI: 10.1021/acs.jproteome.5b00388] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
American ginseng (Panax quinquefolius L.) is one of the most commonly used herbal medicines in the West. It has been reported to possess significant antitumor effects that inhibit the process of carcinogenesis. However, the mechanisms underlying its anticancer effects remain largely unresolved. In this study, we investigated the cancer chemopreventive effects of American ginseng on the progression of high fat (HF) diet-enhanced colorectal carcinogenesis with a genetically engineered Apc(Min/+) mouse model. The metabolic alterations in sera of experimental mice perturbed by HF diet intervention as well as the American ginseng treatment were measured by gas chromatography time-of-flight mass spectrometry (GC-TOFMS) and liquid chromatography time-of-flight mass spectrometry (LC-TOFMS) analysis. American ginseng treatment significantly extended the life span of the Apc(Min/+) mouse. Significant alterations of metabolites involving amino acids, organic acids, fatty acids, and carbohydrates were observed in Apc(Min/+) mouse in sera, which were attenuated by American ginseng treatment and concurrent with the histopathological improvement with significantly reduced tumor initiation, progression and gut inflammation. These metabolic changes suggest that the preventive effect of American ginseng is associated with attenuation of impaired amino acid, carbohydrates, and lipid metabolism. It also appears that American ginseng induced significant metabolic alterations independent of the Apc(Min/+) induced metabolic changes. The significantly altered metabolites induced by American ginseng intervention include arachidonic acid, linolelaidic acid, glutamate, docosahexaenoate, tryptophan, and fructose, all of which are associated with inflammation and oxidation. This suggests that American ginseng exerts the chemopreventive effects by anti-inflammatory and antioxidant mechanisms.
Collapse
Affiliation(s)
- Guoxiang Xie
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, IL 60637
| | - Chunhao Yu
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, IL 60637
| | - Yunping Qiu
- Albert Einstein College of Medicine, Bronx, NY 10461
| | - Xiao-Dong Wen
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, IL 60637
| | - Chun-Feng Zhang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, IL 60637
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, IL 60637
| | - Wei Jia
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813
| |
Collapse
|
17
|
Estaki M, Noble EG. North American ginseng protects against muscle damage and reduces neutrophil infiltration after an acute bout of downhill running in rats. Appl Physiol Nutr Metab 2014; 40:116-21. [PMID: 25531801 DOI: 10.1139/apnm-2014-0331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Eccentric muscle contractions such as those experienced during downhill running are associated with inflammation, delayed-onset of muscle soreness, myofiber damage, and various functional deficits. North American ginseng (Panax quinquefolius L.) has been reported to possess anti-inflammatory properties and thus may offset some of this exercise-induced damage. Hence, we tested the hypothesis that intervention with North American ginseng would reduce eccentric exercise-induced muscle damage and inflammation. Male Wistar rats were fed (300 mg/(kg·day)(-1)) of either an alcohol (AL) or aqueous (AQ) extract of North American ginseng for 14 days before a single bout of downhill running and were compared with matching nonexercised (C) groups. Plasma creatine kinase levels were significantly reduced in both ginseng treated groups compared with the C group that received a water placebo (p < 0.002). Further, the AQ but not AL group also showed attenuated morphological signs of damage (hemotoxylin and eosin) as well as reduced levels of infiltrating neutrophils (HIS48) in the soleus muscle (p < 0.001). In summary, supplementation with an AQ but not AL extract of North American ginseng was able to reduce eccentric exercise-induced muscle damage and inflammation.
Collapse
Affiliation(s)
- Mehrbod Estaki
- a School of Kinesiology, The University of Western Ontario, London, ON N6A 3K7, Canada
| | | |
Collapse
|
18
|
Cho SH. Red ginseng for atopic dermatitis. World J Dermatol 2014; 3:58-63. [DOI: 10.5314/wjd.v3.i3.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/14/2014] [Accepted: 07/29/2014] [Indexed: 02/06/2023] Open
Abstract
Red ginseng is known for its significant biological activities which include anti-inflammation. Red ginseng may be used for the management and prevention of atopic dermatitis based on its effect on an atopic dermatitis animal model. More therapeutic efficacies other than atopic dermatitis are also reviewed briefly.
Collapse
|
19
|
American ginseng attenuates azoxymethane/dextran sodium sulfate-induced colon carcinogenesis in mice. J Ginseng Res 2014; 39:14-21. [PMID: 25535472 PMCID: PMC4268560 DOI: 10.1016/j.jgr.2014.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Colorectal cancer is a leading cause of cancer-related death, and inflammatory bowel disease is a risk factor for this malignancy. We previously reported colon cancer chemoprevention potential using American ginseng (AG) in a xenograft mice model. However, the nude mouse model is not a gut-specific colon carcinogenesis animal model. METHODS In this study, an experimental colitis and colitis-associated colorectal carcinogenesis mouse model, chemically induced by azoxymethane/dextran sodium sulfate (DSS) was established and the effects of oral AG were evaluated. The contents of representative ginseng saponins in the extract were determined. RESULTS AG significantly reduced experimental colitis measured by the disease activity index scores. This suppression of the experimental colitis was not only evident during DSS treatment, but also very obvious after the cessation of DSS, suggesting that the ginseng significantly promoted recovery from the colitis. Consistent with the anti-inflammation data, we showed that ginseng very significantly attenuated azoxymethane/DSS-induced colon carcinogenesis by reducing the colon tumor number and tumor load. The ginseng also effectively suppressed DSS-induced proinflammatory cytokines activation using an enzyme-linked immunosorbent assay array, in which 12 proinflammatory cytokine levels were assessed, and this effect was supported subsequently by real-time polymerase chain reaction data. CONCLUSION AG, as a candidate of botanical-based colon cancer chemoprevention, should be further investigated for its potential clinical utility.
Collapse
|
20
|
Oh G, Yoo SW, Jung Y, Ryu YM, Park Y, Kim SY, Kim KH, Kim S, Myung SJ, Chung E. Intravital imaging of mouse colonic adenoma using MMP-based molecular probes with multi-channel fluorescence endoscopy. BIOMEDICAL OPTICS EXPRESS 2014; 5:1677-89. [PMID: 24877024 PMCID: PMC4026906 DOI: 10.1364/boe.5.001677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/19/2014] [Accepted: 04/21/2014] [Indexed: 05/18/2023]
Abstract
Intravital imaging has provided molecular, cellular and anatomical insight into the study of tumor. Early detection and treatment of gastrointestinal (GI) diseases can be enhanced with specific molecular markers and endoscopic imaging modalities. We present a wide-field multi-channel fluorescence endoscope to screen GI tract for colon cancer using multiple molecular probes targeting matrix metalloproteinases (MMP) conjugated with quantum dots (QD) in AOM/DSS mouse model. MMP9 and MMP14 antibody (Ab)-QD conjugates demonstrate specific binding to colonic adenoma. The average target-to-background (T/B) ratios are 2.10 ± 0.28 and 1.78 ± 0.18 for MMP14 Ab-QD and MMP9 Ab-QD, respectively. The overlap between the two molecular probes is 67.7 ± 8.4%. The presence of false negative indicates that even more number of targeting could increase the sensitivity of overall detection given heterogeneous molecular expression in tumors. Our approach indicates potential for the screening of small or flat lesions that are precancerous.
Collapse
Affiliation(s)
- Gyungseok Oh
- School of Mechatronics, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Su Woong Yoo
- Department of Medical System Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Yebin Jung
- Department of Chemistry, Pohang University of Science and Technology, Pohang, South Korea
| | - Yeon-Mi Ryu
- Asan Institute for Life sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Youngrong Park
- Department of Chemistry, Pohang University of Science and Technology, Pohang, South Korea
| | - Sang-Yeob Kim
- Asan Institute for Life sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Deparment of Medicine, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ki Hean Kim
- Division of Integrative Biosciences and Biotechnology and Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Sungjee Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang, South Korea
| | - Seung-Jae Myung
- Asan Institute for Life sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Department of Gastroenterology, Asan Medical Center,University of Ulsan College of Medicine, Seoul, South Korea
| | - Euiheon Chung
- School of Mechatronics, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Department of Medical System Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|
21
|
Poudyal D, Cui X, Le PM, Hofseth AB, Windust A, Nagarkatti M, Nagarkatti PS, Schetter AJ, Harris CC, Hofseth LJ. A key role of microRNA-29b for the suppression of colon cancer cell migration by American ginseng. PLoS One 2013; 8:e75034. [PMID: 24130681 PMCID: PMC3794036 DOI: 10.1371/journal.pone.0075034] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/07/2013] [Indexed: 11/29/2022] Open
Abstract
Metastasis of colon cancer cells increases the risk of colon cancer mortality. We have recently shown that American ginseng prevents colon cancer, and a Hexane extract of American Ginseng (HAG) has particularly potent anti-inflammatory and anti-cancer properties. Dysregulated microRNA (miR) expression has been observed in several disease conditions including colon cancer. Using global miR expression profiling, we observed increased miR-29b in colon cancer cells following exposure to HAG. Since miR-29b plays a role in regulating the migration of cancer cells, we hypothesized that HAG induces miR-29b expression to target matrix metalloproteinase-2 (MMP-2) thereby suppressing the migration of colon cancer cells. Results are consistent with this hypothesis. Our study supports the understanding that targeting MMP-2 by miR-29b is a mechanism by which HAG suppresses the migration of colon cancer cells.
Collapse
Affiliation(s)
- Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
| | - Xiangli Cui
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
- Shanxi Medical University, Shanxi, China
| | - Phuong Mai Le
- Institute for National Measurement Standards, National Research Council, Ottawa, Canada
| | - Anne B. Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
| | - Anthony Windust
- Institute for National Measurement Standards, National Research Council, Ottawa, Canada
| | - Mitzi Nagarkatti
- School of Medicine, University of South Carolina, Columbia, South Carolina, United States of America
| | - Prakash S. Nagarkatti
- School of Medicine, University of South Carolina, Columbia, South Carolina, United States of America
| | - Aaron J. Schetter
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lorne J. Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
22
|
Sofowora A, Ogunbodede E, Onayade A. The role and place of medicinal plants in the strategies for disease prevention. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2013; 10:210-29. [PMID: 24311829 PMCID: PMC3847409 DOI: 10.4314/ajtcam.v10i5.2] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Medicinal plants have been used in healthcare since time immemorial. Studies have been carried out globally to verify their efficacy and some of the findings have led to the production of plant-based medicines. The global market value of medicinal plant products exceeds $100 billion per annum. This paper discusses the role, contributions and usefulness of medicinal plants in tackling the diseases of public health importance, with particular emphasis on the current strategic approaches to disease prevention. A comparison is drawn between the 'whole population' and 'high-risk' strategies. The usefulness of the common-factor approach as a method of engaging other health promoters in propagating the ideals of medicinal plants is highlighted. The place of medicinal plants in preventing common diseases is further examined under the five core principles of the Primary Health Care (PHC) approach. Medicinal plants play vital roles in disease prevention and their promotion and use fit into all existing prevention strategies. However, conscious efforts need to be made to properly identify, recognise and position medicinal plants in the design and implementation of these strategies. These approaches present interesting and emerging perspectives in the field of medicinal plants. Recommendations are proposed for strategising the future role and place for medicinal plants in disease prevention.
Collapse
Affiliation(s)
- Abayomi Sofowora
- C/O Department of Pharmacognosy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | | | | |
Collapse
|
23
|
Kim SK, Park JH. Trends in ginseng research in 2010. J Ginseng Res 2013; 35:389-98. [PMID: 23717084 PMCID: PMC3659559 DOI: 10.5142/jgr.2011.35.4.389] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 11/22/2011] [Accepted: 11/22/2011] [Indexed: 11/30/2022] Open
Abstract
A total of 470 papers directly related to research on the Panax species were retrieved by performing internet searches with the keywords Panax and ginseng as the search terms. The publications were categorized as follows: 399 research articles, 30 reviews, 30 meeting abstracts, 7 proceedings, and 4 letters. The majority of these publications were published by scientists from Korea (35.7%), China (32.3%), and the USA (11.3%). Scientists from a total of 29 nations were actively involved in conducting ginseng research. A total of 43.6% of the publications were categorized as pharmacodynamic studies. The effects of ginseng on cerebrovascular function and cancer were the two most common topics considered in the pharmacodynamic studies. More than half of the ginseng studies assessed the use of P. ginseng. A total of 23 countries participated in studies specifically related to P. ginseng, and more than 80% of these studies originated from Korea and China. A total of 50 topics within the pharmacodynamics category were examined in association with the use of P. ginseng.
Collapse
Affiliation(s)
- Si-Kwan Kim
- Department of Life Sciences, College of Biomedical and Health Sciences, Konkuk University, Chungju 380-701, Korea
| | | |
Collapse
|
24
|
Wei Q, Jiang H, Baker A, Dodge LK, Gerard M, Young MR, Toledano MB, Colburn NH. Loss of sulfiredoxin renders mice resistant to azoxymethane/dextran sulfate sodium-induced colon carcinogenesis. Carcinogenesis 2013; 34:1403-10. [PMID: 23393226 DOI: 10.1093/carcin/bgt059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sulfiredoxin (Srx) is the enzyme that reduces the hyperoxidized inactive form of peroxiredoxins. To study the function of Srx in carcinogenesis in vivo, we tested whether loss of Srx protects mice from cancer development. Srx null mice were generated and colon carcinogenesis was induced by an azoxymethane (AOM) and dextran sulfate sodium (DSS) protocol. Compared with either wild-type (Wt) or heterozygotes, Srx(-/-) mice had significantly reduced rates in both tumor multiplicity and volume. Mechanistic studies reveal that loss of Srx did not alter tumor cell proliferation; however, increased apoptosis and decreased inflammatory cell infiltration were obvious in tumors from Srx null mice compared with those from Wt control. In addition to the AOM/DSS model, examination of Srx expression in human reveals a tissue-specific expression pattern. Srx expression was also demonstrated in tumors from colorectal cancer patients and the levels of expression were associated with patients' clinic stages. These data provide the first in vivo evidence that loss of Srx renders mice resistant to AOM/DSS-induced colon carcinogenesis, suggesting that Srx has a critical oncogenic role in cancer development, and Srx may be used as a marker for human colon cancer pathogenicity.
Collapse
Affiliation(s)
- Qiou Wei
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Poudyal D, Le PM, Davis T, Hofseth AB, Chumanevich A, Chumanevich AA, Wargovich MJ, Nagarkatti M, Nagarkatti PS, Windust A, Hofseth LJ. Hexane Fraction of American Ginseng Suppresses Colitis and Colon Cancer—Response. Cancer Prev Res (Phila) 2012. [DOI: 10.1158/1940-6207.capr-12-0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Deepak Poudyal
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Phuong Mai Le
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Tia Davis
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Anne B. Hofseth
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Alena Chumanevich
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Alexander A. Chumanevich
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Michael J. Wargovich
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Mitzi Nagarkatti
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Prakash S. Nagarkatti
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Anthony Windust
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Lorne J. Hofseth
- Authors' Affiliations: 1Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina and Medical University of South Carolina; 2Department of Biological Sciences, 3School of Medicine, University of South Carolina, Columbia, South Carolina; 4Institute for National Measurement Standards, National Research Council, Ottawa, Ontario, Canada; and 5Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
26
|
Poudyal D, Le PM, Davis T, Hofseth AB, Chumanevich A, Chumanevich AA, Wargovich MJ, Nagarkatti M, Nagarkatti PS, Windust A, Hofseth LJ. A hexane fraction of American ginseng suppresses mouse colitis and associated colon cancer: anti-inflammatory and proapoptotic mechanisms. Cancer Prev Res (Phila) 2012; 5:685-96. [PMID: 22293630 DOI: 10.1158/1940-6207.capr-11-0421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ulcerative colitis is a chronic inflammatory condition associated with a high colon cancer risk. We have previously reported that American ginseng extract significantly reduced the inflammatory parameters of chemically induced colitis. The aim of this study was to further delineate the components of American ginseng that suppress colitis and prevent colon cancer. Among five different fractions of American ginseng (butanol, hexane, ethylacetate, dichloromethane, and water), a hexane fraction has particularly potent antioxidant and proapoptotic properties. The effects of this fraction were shown in a mouse macrophage cell line (ANA-1 cells), in a human lymphoblastoid cell line (TK6), and in an ex vivo model (CD4(+)/CD25(-) primary effector T cells). A key in vivo finding was that compared with the whole American ginseng extract, the hexane fraction of American ginseng was more potent in treating colitis in a dextran sodium sulfate (DSS) mouse model, as well as suppressing azoxymethane/DSS-induced colon cancer. Furthermore, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) labeling of inflammatory cells within the colonic mesenteric lymph nodes was elevated in mice consuming DSS + the hexane fraction of American ginseng. Results are consistent with our in vitro data and with the hypothesis that the hexane fraction of American ginseng has anti-inflammatory properties and drives inflammatory cell apoptosis in vivo, providing a mechanism by which this fraction protects from colitis in this DSS mouse model. This study moves us closer to understanding the molecular components of American ginseng that suppress colitis and prevent colon cancer associated with colitis.
Collapse
Affiliation(s)
- Deepak Poudyal
- Department of Biomedical and Pharmaceutical Sciences, South Carolina College of Pharmacy, University of South Carolina, 770 Sumter St., Coker Life Sciences, Columbia, SC 29208, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Potential agents for cancer and obesity treatment with herbal medicines from the green garden. BIOTECHNOL BIOPROC E 2011. [DOI: 10.1007/s12257-011-0215-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Dougherty U, Mustafi R, Wang Y, Musch MW, Wang CZ, Konda VJ, Kulkarni A, Hart J, Dawson G, Kim KE, Yuan CS, Chang EB, Bissonnette M. American ginseng suppresses Western diet-promoted tumorigenesis in model of inflammation-associated colon cancer: role of EGFR. Altern Ther Health Med 2011; 11:111. [PMID: 22070864 PMCID: PMC3227598 DOI: 10.1186/1472-6882-11-111] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 11/09/2011] [Indexed: 02/06/2023]
Abstract
Background Western diets increase colon cancer risk. Epidemiological evidence and experimental studies suggest that ginseng can inhibit colon cancer development. In this study we asked if ginseng could inhibit Western diet (20% fat) promoted colonic tumorigenesis and if compound K, a microbial metabolite of ginseng could suppress colon cancer xenograft growth. Methods Mice were initiated with azoxymethane (AOM) and, two weeks later fed a Western diet (WD, 20% fat) alone, or WD supplemented with 250-ppm ginseng. After 1 wk, mice received 2.5% dextran sulfate sodium (DSS) for 5 days and were sacrificed 12 wks after AOM. Tumors were harvested and cell proliferation measured by Ki67 staining and apoptosis by TUNEL assay. Levels of EGF-related signaling molecules and apoptosis regulators were determined by Western blotting. Anti-tumor effects of intraperitoneal compound K were examined using a tumor xenograft model and compound K absorption measured following oral ginseng gavage by UPLC-mass spectrometry. Effects of dietary ginseng on microbial diversity were measured by analysis of bacterial 16S rRNA. Results Ginseng significantly inhibited colonic inflammation and tumorigenesis and concomitantly reduced proliferation and increased apoptosis. The EGFR cascade was up-regulated in colonic tumors and ginseng significantly reduced EGFR and ErbB2 activation and Cox-2 expression. Dietary ginseng altered colonic microbial diversity, and bacterial suppression with metronidazole reduced serum compound K following ginseng gavage. Furthermore, compound K significantly inhibited tumor xenograft growth. Conclusions Ginseng inhibited colonic inflammation and tumorigenesis promoted by Western diet. We speculate that the ginseng metabolite compound K contributes to the chemopreventive effects of this agent in colonic tumorigenesis.
Collapse
|
29
|
Hwang SY, Ahn SH. Biological Activities and Cell Proliferation effects of Red Ginseng Ethanol Extracts. J Pharmacopuncture 2011. [DOI: 10.3831/kpi.2011.14.3.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|