1
|
Li X, Owen LA, Taylor KD, Ostmo S, Chen YDI, Coyner AS, Sonmez K, Hartnett ME, Guo X, Ipp E, Roll K, Genter P, Chan RVP, DeAngelis MM, Chiang MF, Campbell JP, Rotter JI. Genome-wide association identifies novel ROP risk loci in a multiethnic cohort. Commun Biol 2024; 7:107. [PMID: 38233474 PMCID: PMC10794688 DOI: 10.1038/s42003-023-05743-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024] Open
Abstract
We conducted a genome-wide association study (GWAS) in a multiethnic cohort of 920 at-risk infants for retinopathy of prematurity (ROP), a major cause of childhood blindness, identifying 1 locus at genome-wide significance level (p < 5×10-8) and 9 with significance of p < 5×10-6 for ROP ≥ stage 3. The most significant locus, rs2058019, reached genome-wide significance within the full multiethnic cohort (p = 4.96×10-9); Hispanic and European Ancestry infants driving the association. The lead single nucleotide polymorphism (SNP) falls in an intronic region within the Glioma-associated oncogene family zinc finger 3 (GLI3) gene. Relevance for GLI3 and other top-associated genes to human ocular disease was substantiated through in-silico extension analyses, genetic risk score analysis and expression profiling in human donor eye tissues. Thus, we identify a novel locus at GLI3 with relevance to retinal biology, supporting genetic susceptibilities for ROP risk with possible variability by race and ethnicity.
Collapse
Affiliation(s)
- Xiaohui Li
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leah A Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA.
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA.
- Department of Ophthalmology, University at Buffalo the State University of New York, Buffalo, NY, USA.
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Susan Ostmo
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Aaron S Coyner
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kemal Sonmez
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eli Ipp
- Division of Endocrinology and Metabolism, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kathryn Roll
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pauline Genter
- Division of Endocrinology and Metabolism, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - R V Paul Chan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Margaret M DeAngelis
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Ophthalmology, University at Buffalo the State University of New York, Buffalo, NY, USA
- Department of Biochemistry; Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
- Department of Neuroscience; Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
- Department of Genetics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/State University of New York (SUNY), Buffalo, NY, USA
| | - Michael F Chiang
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
- National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - J Peter Campbell
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA.
| |
Collapse
|
2
|
Park HR, Lee SE, Yi Y, Moon S, Yoon H, Kang CW, Kim J, Park YS. Integrated analysis of miRNA and mRNA expression profiles in diabetic mouse kidney treated to Korean Red Ginseng. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00272-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
3
|
Yang Z, Dan Wang, Li Y, Zhou X, Liu T, Shi C, Li R, Zhang Y, Zhang J, Yan J, Zhu X, Li Y, Gong M, Wang C, Yuan C, Cui Y, Wu X. Untargeted metabolomics analysis of the anti-diabetic effect of Red ginseng extract in Type 2 diabetes Mellitus rats based on UHPLC-MS/MS. Biomed Pharmacother 2022; 146:112495. [PMID: 34891123 DOI: 10.1016/j.biopha.2021.112495] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 11/27/2022] Open
Abstract
Red ginseng is a traditional Chinese herbal medicine that has long been used to treat diabetes, and its blood sugar-lowering activity has been confirmed. However, the mechanism of action of red ginseng on type 2 diabetes mellitus (T2DM) at the metabolic level is still unclear. The purpose of this study is to investigate the effect of red ginseng extract in the treatment of T2DM rats based on untargeted metabolomics. The rat model of T2DM was induced by a high-fat diet (HFD) combined with streptozotocin (STZ), and serum samples were collected after four weeks of treatment. The ultra-high-performance liquid chromatography coupled with Q Exactive HF-X Mass Spectrometer was used to analyze the level of metabolites in serum to evaluate the differences in metabolic levels between different groups. The results of biochemical analysis showed that red ginseng extract intervention significantly improved the levels of total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), serum glucose (GLU), and fasting insulin (FINS) after four weeks. Orthogonal partial least squares discriminant analysis was used to study the overall changes of rat metabolomics. After the intervention of red ginseng extract, 50 biomarkers showed a callback trend. Metabolic pathway enrichment analysis showed that the regulated pathways were D-arginine and D-ornithine metabolism, D-glutamine and D-glutamate metabolism, taurine and hypotaurine metabolism, arginine biosynthesis, and tryptophan metabolism. Generally, the results demonstrated that red ginseng extract had beneficial effects on T2DM, which could be mediated via ameliorating the metabolic disorders.
Collapse
MESH Headings
- Amino Acids/metabolism
- Animals
- Biomarkers/blood
- Chromatography, High Pressure Liquid
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Lipid Metabolism
- Male
- Metabolic Networks and Pathways/drug effects
- Metabolomics
- Panax
- Pancreas/drug effects
- Pancreas/pathology
- Phytotherapy
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Rats, Sprague-Dawley
- Tandem Mass Spectrometry
- Rats
Collapse
Affiliation(s)
- Zijun Yang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Dan Wang
- Department of Pharmacy, Chu Hisen-I Memorial Hospital, Tianjin Medical University, Tianjin 300134, China
| | - Yuanyuan Li
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Department of Pharmacy, Tianjin Fourth Central Hospital, Tianjin 300140, China
| | - Xinfeng Zhou
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Tiantian Liu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Chang Shi
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Rongshan Li
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yanwen Zhang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jun Zhang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiuxing Yan
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xuehui Zhu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University, Tianjin 300052, China
| | - Min Gong
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Chongzhi Wang
- Tang Center for Herbal Medicine Research, University of Chicago, Illinois 60637, USA
| | - Chunsu Yuan
- Tang Center for Herbal Medicine Research, University of Chicago, Illinois 60637, USA
| | - Yan Cui
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Xiaohui Wu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
4
|
Liu Y, Zhang H, Dai X, Zhu R, Chen B, Xia B, Ye Z, Zhao D, Gao S, Orekhov AN, Zhang D, Wang L, Guo S. A comprehensive review on the phytochemistry, pharmacokinetics, and antidiabetic effect of Ginseng. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153717. [PMID: 34583224 DOI: 10.1016/j.phymed.2021.153717] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/08/2021] [Accepted: 08/15/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Radix Ginseng, one of the well-known medicinal herbs, has been used in the management of diabetes and its complications for more than 1000 years. PURPOSE The aim of this review is devoted to summarize the phytochemistry and pharmacokinetics of Ginseng, and provide evidence for the antidiabetic effects of Ginseng and its ingredients as well as the underlying mechanisms involved. METHODS For the purpose of this review, the following databases were consulted: the PubMed Database (https://pubmed.ncbi.nlm.nih.gov), Chinese National Knowledge Infrastructure (http://www.cnki.net), National Science and Technology Library (http://www.nstl.gov.cn/), Wanfang Data (http://www.wanfangdata.com.cn/) and the Web of Science Database (http://apps.webofknowledge.com/). RESULTS Ginseng exhibits glucose-lowering effects in different diabetic animal models. In addition, Ginseng may prevent the development of diabetic complications, including liver, pancreas, adipose tissue, skeletal muscle, nephropathy, cardiomyopathy, retinopathy, atherosclerosis and others. The main ingredients of Ginseng include ginsenosides and polysaccharides. The underlying mechanisms whereby this herb exerts antidiabetic activities may be attributed to the regulation of multiple signaling pathways, including IRS1/PI3K/AKT, LKB1/AMPK/FoxO1, AGEs/RAGE, MAPK/ERK, NF-κB, PPARδ/STAT3, cAMP/PKA/CERB and HIF-1α/VEGF, etc. The pharmacokinetic profiles of ginsenosides provide valuable information on therapeutic efficacy of Ginseng in diabetes. Although Ginseng is well-tolerated, dietary consumption of this herb should follow the doctors' advice. CONCLUSION Ginseng may offer an alternative strategy in protection against diabetes and its complications through the regulations of the multi-targets via various signaling pathways. Efforts to understand the underlying mechanisms with strictly-controlled animal models, combined with well-designed clinical trials and pharmacokinetic evaluation, will be important subjects of the further investigations and weigh in translational value of this herb in diabetes management.
Collapse
Affiliation(s)
- Yage Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hao Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xuan Dai
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ruyuan Zhu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Beibei Chen
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Bingke Xia
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zimengwei Ye
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dandan Zhao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Sihua Gao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Lili Wang
- Department of TCM Pharmacology, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Shuzhen Guo
- Department of Scientific Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
5
|
Niu Z, Shi Y, Li J, Qiao S, Du S, Chen L, Tian H, Wei L, Cao H, Wang J, Gao L. Protective effect of rapamycin in models of retinal degeneration. Exp Eye Res 2021; 210:108700. [PMID: 34245755 DOI: 10.1016/j.exer.2021.108700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
Age-related macular degeneration (AMD) is a complex retinal disease with no viable treatment strategy. The causative mechanistic pathway for this disease is not yet clear. Therefore, it is highly warranted to screen effective drugs to treat AMD. Rapamycin are known to inhibit inflammation and has been widely used in the clinic as an immunosuppressant. This study aimed to investigate the protective effect of rapamycin on the AMD retinal degeneration model. The AMD models were established by injection of 35 mg/kg sodium iodate (NaIO3) into the tail vein. Then the treated mice intraperitoneally received rapamycin (2 mg/kg) once a day. The histomorphological analysis showed that rapamycin could inhibit retinal structure damage and apoptosis. Experiments revealed that rapamycin significantly attenuated inflammatory response and oxidative stress. Our experimental results demonstrated that rapamycin has protected the retinal against degeneration induced by NaIO3. The therapeutic effect was more significant after 7 days of treatment. Therefore, our study potentially provides a powerful experimental support for the treatment of AMD.
Collapse
Affiliation(s)
- Zhanyu Niu
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jiande Li
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Shufan Qiao
- Colloge of Life Sciences, Northwest Normal University, Lanzhou, 730000, China.
| | - Shaobo Du
- School of Stomatology of Lanzhou University, Lanzhou, 730000, China.
| | - Linchi Chen
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Li Wei
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Effect of Korean Red Ginseng through comparative analysis of cardiac gene expression in db/db mice. J Ginseng Res 2020; 45:450-455. [PMID: 34025138 PMCID: PMC8134846 DOI: 10.1016/j.jgr.2020.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/12/2020] [Accepted: 06/21/2020] [Indexed: 11/22/2022] Open
Abstract
Korean Red Ginseng (KRG) is an herbal oriental medicine known to alleviate cardiovascular dysfunction. To analysis the expression of diabetic cardiac complication-associated genes in db/db mice, we studied the cardiac gene expression following KRG treatment. In result, a total of 585 genes were found to be changed in db/db mice. Among the changed expression, 245 genes were found to 2-fold upregulated, and 340 genes were 2-fold downregulated. In addition, the changed gene expressions were ameliorated by KRG. In conclusion, KRG may be possible to normalize cardiac gene expressions in db/db mice.
Collapse
|
7
|
Ginseng for an eye: effects of ginseng on ocular diseases. J Ginseng Res 2020; 44:1-7. [PMID: 32095091 PMCID: PMC7033367 DOI: 10.1016/j.jgr.2018.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/29/2018] [Accepted: 11/26/2018] [Indexed: 01/01/2023] Open
Abstract
The sense of vision is the primary means by which we gather information from our surroundings, and vision loss, therefore, severely compromises the life of the affected individuals, their families, and society. Loss of vision becomes more frequent with age, and diabetic retinopathy, age-related macular degeneration, cataracts, and glaucoma are the major causes of vision impairment. To find active pharmacological compounds that might prevent or ameliorate the vision-threatening eye diseases, numerous studies have been performed, and some botanical compounds, including those extracted from ginseng, have been shown to possess beneficial effects in the treatment or prevention of common ocular diseases. In this review, we summarize the recent reports investigating the therapeutic effects of ginseng and ginsenosides on diverse ocular diseases and discuss their therapeutic potential.
Collapse
|
8
|
Park SJ, Lee D, Kim D, Lee M, In G, Han ST, Kim SW, Lee MH, Kim OK, Lee J. The non-saponin fraction of Korean Red Ginseng (KGC05P0) decreases glucose uptake and transport in vitro and modulates glucose production via down-regulation of the PI3K/AKT pathway in vivo. J Ginseng Res 2019; 44:362-372. [PMID: 32148419 PMCID: PMC7031776 DOI: 10.1016/j.jgr.2019.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/08/2019] [Accepted: 12/10/2019] [Indexed: 01/26/2023] Open
Abstract
Background The non-saponin fraction of Korean Red Ginseng has been reported to have many biological activities. However, the effect of this fraction on anti-diabetic activity has not been elucidated in detail. In this study, we investigated the effects of KGC05P0, a non-saponin fraction of Korean Red Ginseng, on anti-diabetic activity in vitro and in vivo. Methods We measured the inhibition of commercially obtained α-glucosidase and α-amylase activities in vitro and measured the glucose uptake and transport rate in Caco-2 cells. C57BL/6J mice and C57BLKS/Jdb/db (diabetic) mice were fed diets with or without KGC05P0 for eight weeks. To perform the experiments, the groups were divided as follows: normal control (C57BL/6J mice), db/db control (C57BLKS/Jdb/db mice), positive control (inulin 400 mg/kg b.w.), low (KGC05P0 100 mg/kg b.w.), medium (KGC05P0 200 mg/kg b.w.), and high (KGC05P0 400 mg/kg b.w.). Results KGC05P0 inhibited α-glucosidase and α-amylase activities in vitro, and decreased glucose uptake and transport rate in Caco-2 cells. In addition, KGC05P0 regulated fasting glucose level, glucose tolerance, insulin, HbA1c, carbonyl contents, and proinflammatory cytokines in blood from diabetic mice and significantly reduced urinary glucose excretion levels. Moreover, we found that KGC05P0 regulated glucose production by down-regulation of the PI3K/AKT pathway, which inhibited gluconeogenesis. Conclusion Our study thereby demonstrated that KGC05P0 exerted anti-diabetic effects through inhibition of glucose absorption and the PI3K/AKT pathway in in vitro and in vivo models of diabetes. Our results suggest that KGC05P0 could be developed as a complementary food to help prevent T2DM and its complications.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Department of Medical Nutrition, Kyung Hee University, Yongin, Gwangju, Republic of Korea
| | - Dasom Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin, Gwangju, Republic of Korea
| | - Dakyung Kim
- Department of Medical Nutrition, Kyung Hee University, Yongin, Gwangju, Republic of Korea
| | - Minhee Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin, Gwangju, Republic of Korea
| | - Gyo In
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon, Gwangju, Republic of Korea
| | - Sung-Tai Han
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon, Gwangju, Republic of Korea
| | - Sung Won Kim
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon, Gwangju, Republic of Korea
| | - Mi-Hyang Lee
- Korea Ginseng Corporation Research Institute, Korea Ginseng Corporation, Daejeon, Gwangju, Republic of Korea
| | - Ok-Kyung Kim
- Division of Food and Nutrition and Research Institute for Human Ecology, Chonnam National University, Gwangju, Republic of Korea
| | - Jeongmin Lee
- Department of Medical Nutrition, Kyung Hee University, Yongin, Gwangju, Republic of Korea
| |
Collapse
|
9
|
Review of Ginseng Anti-Diabetic Studies. Molecules 2019; 24:molecules24244501. [PMID: 31835292 PMCID: PMC6943541 DOI: 10.3390/molecules24244501] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/03/2019] [Accepted: 12/06/2019] [Indexed: 12/22/2022] Open
Abstract
Ginseng is one of the most valuable and commonly used Chinese medicines not only in ancient China but also worldwide. Ginsenosides, also known as saponins or triterpenoids, are thought to be responsible for the beneficial effects of ginseng. In this review, we summarize recent publications on anti-diabetic studies of ginseng extracts and ginsenosides in cells, animals, and humans. It seems that the anti-diabetic effect of ginseng is positive for type 2 diabetic patients but has no significant impact on prediabetes or healthy adults. Regulation of insulin secretion, glucose uptake, anti-oxidative stress, and anti-inflammatory pathways may be the mechanisms involved with ginseng's anti-diabetic effects. Taken together, this summary provides evidence for the anti-diabetes effects of ginseng extracts and ginsenosides as well as the underlying mechanisms of their impact on diabetes.
Collapse
|
10
|
Iacono G, Massoni-Badosa R, Heyn H. Single-cell transcriptomics unveils gene regulatory network plasticity. Genome Biol 2019; 20:110. [PMID: 31159854 PMCID: PMC6547541 DOI: 10.1186/s13059-019-1713-4] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) plays a pivotal role in our understanding of cellular heterogeneity. Current analytical workflows are driven by categorizing principles that consider cells as individual entities and classify them into complex taxonomies. RESULTS We devise a conceptually different computational framework based on a holistic view, where single-cell datasets are used to infer global, large-scale regulatory networks. We develop correlation metrics that are specifically tailored to single-cell data, and then generate, validate, and interpret single-cell-derived regulatory networks from organs and perturbed systems, such as diabetes and Alzheimer's disease. Using tools from graph theory, we compute an unbiased quantification of a gene's biological relevance and accurately pinpoint key players in organ function and drivers of diseases. CONCLUSIONS Our approach detects multiple latent regulatory changes that are invisible to single-cell workflows based on clustering or differential expression analysis, significantly broadening the biological insights that can be obtained with this leading technology.
Collapse
Affiliation(s)
- Giovanni Iacono
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain.
| | - Ramon Massoni-Badosa
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, 08028, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
11
|
Lee YY, Seo HW, Kyung JS, Hyun SH, Han BC, Park S, So SH, Lee SH, Yi EC. Proteomic studies of putative molecular signatures for biological effects by Korean Red Ginseng. J Ginseng Res 2019; 43:666-675. [PMID: 31700262 PMCID: PMC6823813 DOI: 10.1016/j.jgr.2019.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 04/17/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022] Open
Abstract
Background Korean Red Ginseng (KRG) has been widely used as an herbal medicine to normalize and strengthen body functions. Although many researchers have focused on the biological effects of KRG, more studies on the action mechanism of red ginseng are still needed. Previously, we investigated the proteomic changes of the rat spleen while searching for molecular signatures and the action mechanism of KRG. The proteomic analysis revealed that differentially expressed proteins (DEPs) were involved in the increased immune response and phagocytosis. The aim of this study was to evaluate the biological activities of KRG, especially the immune-enhancing response of KRG. Methods Rats were divided into 4 groups: 0 (control group), 500, 1000, and 2000 mg/kg administration of KRG powder for 6 weeks, respectively. Isobaric tags for relative and absolute quantitation was performed with Q-Exactive LC-MS/MS to compare associated proteins between the groups. The putative DEPs were identified by a current UniProt rat protein database search and by the Gene Ontology annotations. Results The DEPs appear to increase the innate and acquired immunity as well as immune cell movement. These results suggest that KRG can stimulate immune responses. This analysis refined our targets of interest to include the potential functions of KRG. Furthermore, we validated the potential molecular targets of the functions, representatively LCN2, CRAMP, and HLA-DQB1, by Western blotting. Conclusion These results may provide molecular signature candidates to elucidate the mechanisms of the immune response by KRG. Here, we demonstrate a strategy of tissue proteomics for the discovery of the molecular function of KRG.
Collapse
Affiliation(s)
- Yong Yook Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Hwi Won Seo
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Jong-Su Kyung
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Sun Hee Hyun
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Byung Cheol Han
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Songhee Park
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Seung Ho So
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Seung Ho Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Dong C, Liu P, Wang H, Dong M, Li G, Li Y. Ginsenoside Rb1 attenuates diabetic retinopathy in streptozotocin-induced diabetic rats1. Acta Cir Bras 2019; 34:e201900201. [PMID: 30843934 PMCID: PMC6585913 DOI: 10.1590/s0102-8650201900201] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/15/2019] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigated the effects of ginsenoside Rb1 on diabetic retinopathy in streptozotocin-induced diabetic rats. METHODS Diabetes was induced by a single intraperitoneal injection of streptozotocin (80 mg/kg) in male Wistar rats. Ginsenoside Rb1 (20, 40 mg/kg) was injected (i.p.) once a day for 4 weeks. Then, using fundus photography, the diameter and vascular permeability of retinal vessels were investigated. Retinal histopathology was undertaken. Contents of malondialdehyde (MDA) and glutathione (GSH) in retinas were assayed. Levels of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione cysteine ligase catalytic subunit (GCLC), and glutathione cysteine ligase modulatory subunit (GCLM) were measured. RESULTS Treatment with ginsenoside Rb1 attenuated the diabetes-induced increase in the diameter of retinal blood vessels. Ginsenoside Rb1 reduced extravasation of Evans Blue dye from retinal blood vessels. Ginsenoside Rb1 partially inhibited the increase in MDA content and decrease in GSH level in rat retinas. Nrf2 levels in the nuclei of retinal cells and expression of GCLC and GCLM were increased significantly in rats treated with ginsenoside Rb1. CONCLUSION These findings suggest that ginsenoside Rb1 can attenuate diabetic retinopathy by regulating the antioxidative function in rat retinas.
Collapse
Affiliation(s)
- Changxia Dong
- Master, Department of Ophthalmology, Yantai Yuhuangding Hospital, P.R. China. Acquisition of data, manuscript writing
| | - Peng Liu
- Master, Department of Ophthalmology, Yantai Yuhuangding Hospital, P.R. China. Manuscript writing
| | - Huaizhou Wang
- Master, Department of Anesthesiology, Yantai Stomatological Hospital, P.R. China. Acquisition of data
| | - Mei Dong
- Master, Department of Ophthalmology, Yantai Yuhuangding Hospital, P.R. China. Analysis and interpretation of data
| | - Guangxin Li
- Master, Department of Ophthalmology, Yantai Yuhuangding Hospital, P.R. China. Analysis and interpretation of data
| | - Yuanbin Li
- MD, Department of Ophthalmology, Yantai Yuhuangding Hospital, P.R. China. Conception and design of the study, critical revision, final approval
| |
Collapse
|
13
|
Jung E, Kim CS, Jung W, Park SB, Pyo MK, Kim J. Ginseng Extract Modified by Pectin Lyase Inhibits Retinal Vascular Injury and Blood-Retinal Barrier Breakage in a Rat Model of Diabetes. J Med Food 2019; 22:337-343. [PMID: 30785359 DOI: 10.1089/jmf.2018.4256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
GS-E3D is an enzymatically modified ginseng extract by pectin lyase. In this study, we evaluated the preventive effects of GS-E3D on blood-retinal barrier (BRB) leakage in a rat model of diabetes. To produce diabetes, rats were injected with streptozotocin. GS-E3D was orally gavaged at 25, 50, and 100 mg/kg body weight for 6 weeks. We then compared the effect of GS-E3D with that of an unmodified ginseng extract (UGE) on retinal vascular leakage. The administration of GS-E3D significantly blocked diabetes-induced BRB breakdown. Immunofluorescence staining showed that GS-E3D reduced the loss of occludin in diabetic rats. In TUNEL staining, the number of apoptotic retinal microvascular cells was dose dependently decreased by GS-E3D treatment. GS-E3D decreased the accumulations of advanced glycation end products in the retinal vessels. In addition, the inhibition potential of GS-E3D on BRB breakage was stronger compared with UGE. These results indicate that GS-E3D could be a beneficial treatment option for preventing diabetes-induced retinal vascular injury.
Collapse
Affiliation(s)
- Eunsoo Jung
- 1 Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Chan-Sik Kim
- 2 Clinical Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Wookwon Jung
- 3 Department of Oral Pathology, School of Dentistry, Chonbuk National University, Jeonju, South Korea
| | - Su-Bin Park
- 3 Department of Oral Pathology, School of Dentistry, Chonbuk National University, Jeonju, South Korea
| | - Mi-Kyung Pyo
- 4 International Ginseng and Herb Research Institute, Geumsan, South Korea
| | - Junghyun Kim
- 2 Clinical Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.,3 Department of Oral Pathology, School of Dentistry, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
14
|
Karmazyn M, Gan XT. Ginseng for the treatment of diabetes and diabetes-related cardiovascular complications: a discussion of the evidence 1. Can J Physiol Pharmacol 2018; 97:265-276. [PMID: 30395481 DOI: 10.1139/cjpp-2018-0440] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder associated with elevated blood glucose levels due either to insufficient insulin production (type 1 DM) or to insulin resistance (type 2 DM). The incidence of DM around the world continues to rise dramatically with more than 400 million cases reported today. Among the most serious consequences of chronic DM are cardiovascular complications that can have deleterious effects. Although numerous treatment options are available, including both pharmacological and nonpharmacological, there is substantial emerging interest in the use of traditional medicines for the treatment of this condition and its complications. Among these is ginseng, a medicinal herb that belongs to the genus Panax and has been used for thousands of years as a medicinal agent especially in Asian cultures. There is emerging evidence from both animal and clinical studies that ginseng, ginseng constituents including ginsenosides, and ginseng-containing formulations can produce beneficial effects in terms of normalization of blood glucose levels and attenuation of cardiovascular complications through a multiplicity of mechanisms. Although more research is required, ginseng may offer a useful therapy for the treatment of diabetes as well as its complications.
Collapse
|
15
|
Zou HH, Yang PP, Huang TL, Zheng XX, Xu GS. PLK2 Plays an Essential Role in High D-Glucose-Induced Apoptosis, ROS Generation and Inflammation in Podocytes. Sci Rep 2017; 7:4261. [PMID: 28655909 PMCID: PMC5487358 DOI: 10.1038/s41598-017-00686-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/08/2017] [Indexed: 01/15/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious complication of hyperglycemia. Currently, there is no effective therapeutic intervention for DKD. In this study, we sought to provide a set of gene profile in diabetic kidneys. We identified 338 genes altered in diabetes-induced DKD glomeruli, and PLK2 exhibited the most dramatic change. Gene set enrichment analysis (GSEA) indicated multiple signaling pathways are involved DKD pathogenesis. Here, we investigated whether PLK2 contributes to podocyte dysfunction, a characteristic change in the development of DKD. High D-glucose (HDG) significantly increased PLK2 expression in mouse podocytes. Suppressing PLK2 attenuated HDG-induced apoptosis and inflammatory responses both in vitro and in vivo. NAC, an antioxidant reagent, rescued HDG and PLK2 overexpression-induced kidney injuries. In summary, we demonstrated that silencing PLK2 attenuates HDG-induced podocyte apoptosis and inflammation, which may serve as a future therapeutic target in DKD.
Collapse
Affiliation(s)
- Hong-Hong Zou
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, P.R. China
| | - Ping-Ping Yang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, P.R. China
| | - Tian-Lun Huang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, P.R. China
| | - Xiao-Xu Zheng
- Department of Medicine, the George Washington University, Washington, DC20052, USA
| | - Gao-Si Xu
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, P.R. China.
| |
Collapse
|
16
|
Xie J, Wu YY, Zhang TY, Zhang MY, Zhu WW, Gullen EA, Wang ZJ, Cheng YC, Zhang YX. New and bioactive natural products from an endophyte of Panax notoginseng. RSC Adv 2017. [DOI: 10.1039/c7ra07060h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Secondary metabolites with cytotoxic activity, antiviral activity and antimicrobial activity from the endophytic fungi of Panax notoginseng.
Collapse
Affiliation(s)
- Jun Xie
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| | - Ying-Ying Wu
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| | - Tian-Yuan Zhang
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| | - Meng-Yue Zhang
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| | - Wei-Wei Zhu
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| | | | - Zhao-Jie Wang
- Yunnan Provincial Academy of Science and Technology
- Kunming
- People's Republic of China
| | - Yung-Chi Cheng
- Department of Pharmacology
- Yale University School of Medicine
- New Haven
- USA
| | - Yi-Xuan Zhang
- School of Life Science and Biopharmaceutics
- Shenyang Pharmaceutical University
- Shenyang
- People's Republic of China
| |
Collapse
|