1
|
Tao S, Qi Y, Gao J, Yuan H, Wang R, Shen X, Wei G, Peng Z. Ameliorative Effect of Sipunculus nudus Hydrolysate on Cisplatin-Induced Nephrotoxicity by Mitigating Oxidative Stress, Inflammation and Apoptosis. Mar Drugs 2025; 23:100. [PMID: 40137286 PMCID: PMC11943811 DOI: 10.3390/md23030100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 03/27/2025] Open
Abstract
The present study investigated the protective effects and possible mechanisms of an ultrafiltration fraction of Sipunculus nudus hydrolysate (UFSH) on cisplatin-induced kidney damage in a mouse model. The results showed that UFSH significantly attenuated cisplatin-induced nephrotoxicity by inhibiting increases in blood urea nitrogen (BUN) and serum creatinine (SCr). Additionally, UFSH treatment significantly alleviated cisplatin-induced renal histopathological changes, such as significant dilation of renal tubules, cast formation, and tubular cell necrosis, as well as tubulointerstitial fibrosis. Moreover, UFSH decreased cisplatin-induced oxidative stress by increasing the activities of antioxidant enzymes SOD and GSH-Px, while reducing the malondialdehyde (MDA) level in the kidney. Furthermore, UFSH significantly inhibited cisplatin-induced increases in inflammatory cytokines, including Interleukin 1-beta (IL-1β), Interleukin-6 (IL-6), and Tumor necrosis factor-alpha (TNF-α). Western blotting revealed that UFSH inhibited the phosphorylation of the inflammation-associated MAPK/NF-κB signaling pathway, lowered the expression of the apoptosis-related protein Bax, and reversed the reduction in the anti-apoptotic Bcl-2 protein. This investigation demonstrated that UFSH can ameliorate cisplatin-induced nephrotoxicity by mitigating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Susu Tao
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Yi Qi
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Jialong Gao
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
| | - Huafang Yuan
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Ruimin Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Xiaoqin Shen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Gang Wei
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Zhilan Peng
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| |
Collapse
|
2
|
Bai B, Ma Y, Liu D, Zhang Y, Zhang W, Shi R, Zhou Q. DNA damage caused by chemotherapy has duality, and traditional Chinese medicine may be a better choice to reduce its toxicity. Front Pharmacol 2024; 15:1483160. [PMID: 39502534 PMCID: PMC11534686 DOI: 10.3389/fphar.2024.1483160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Background DNA damage induced by chemotherapy has duality. It affects the efficacy of chemotherapy and constrains its application. An increasing number of studies have shown that traditional Chinese medicine (TCM) is highly effective in reducing side-effects induced by chemotherapy due to its natural, non-toxic and many sourced from food. Recent advancements have demonstrated survival rates are improved attributable to effective chemotherapy. DNA damage is the principal mechanism underlying chemotherapy. However, not all instances of DNA damage are beneficial. Chemotherapy induces DNA damage in normal cells, leading to side effects. It affects the efficacy of chemotherapy and constrains its application. Objectives This review aims to summarize the dual nature of DNA damage induced by chemotherapy and explore how TCM can mitigate chemotherapy-induced side effects. Results The review summarized the latest research progress in DNA damage caused by chemotherapy and the effect of alleviating side effects by TCM. It focused on advantages and disadvantages of chemotherapy, the mechanism of drugs and providing insights for rational and effective clinical treatment and serving as a basis for experiment. In this review, we described the mechanisms of DNA damage, associated chemotherapeutics, and their toxicity. Furthermore, we explored Chinese herb that can alleviate chemotherapy-induced side-effects. Conclusion We highlight key mechanisms of DNA damage caused by chemotherapeutics and discuss specific TCM herbs that have shown potential in reducing these side effects. It can provide reference for clinical and basic research.
Collapse
Affiliation(s)
- Bufan Bai
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingrui Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deng Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weihong Zhang
- Breast Surgery Department, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Dongfang Hospital Affiliated to Shanghai Tongji University, Shanghai, China
| |
Collapse
|
3
|
Ji X, Yu L, Han C, Gao H, Cai Y, Li J, He Y, Lu H, Song G, Xue P. Investigating the effects of rare ginsenosides on hyperuricemia and associated sperm damage via nontargeted metabolomics and gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118362. [PMID: 38768838 DOI: 10.1016/j.jep.2024.118362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In ancient times, ginseng was used for hyperuricemia treatment as described in the classic traditional Chinese medical text Shang Han Lun. Recent studies have shown that common ginsenosides and rare ginsenosides (RGS) are the main active compounds in ginseng. RGS have higher activity and are less studied in the treatment of hyperuricemia. AIM OF THE STUDY To determine whether RGS prevents and ameliorates potassium oxonate(PO)-induced hyperuricemia and concomitant spermatozoa damage in mice and the possible underlying mechanisms. MATERIALS AND METHODS Potassium oxonate (PO, 300 mg/kg) induced hyperuricemia in mice via the oral administration of RGS (50, 100, or 200 mg/kg) or allopurinol (ALL, 5 mg/kg) for 35 days. Uric acid (UA) and xanthine oxidase (XO) levels were measured to assess the degree of histopathological damage in the liver, kidney, and testis, and renal creatinine (CRE), urea nitrogen (BUN), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and inflammatory factor (IL-1β) levels were measured to calculate the sperm density. Mechanisms were also explored based on blood and urine metabolomics and the gut microbiota. RESULTS In this study, we demonstrated that RGS containing Rg3, Rk1, Rg6, and Rg5 could reduce serum UA levels, inhibit serum and hepatic XO activity, reduce renal CRE and BUN levels, further restore renal SOD and GSH activities, reduce the accumulation of MDA in the kidneys, and attenuate the production of renal IL-1β. RGS was able to restore sperm density. Metabolomic analysis revealed that RGS improved sphingolipid metabolism, pyrimidine metabolism, and other metabolic pathways. 16S rDNA sequencing revealed that RGS could increase gut microbial diversity, restore the Firmicutes/Bacteroidetes (F/B) ratio, and adjust the intestinal microbial balance. Spearman's correlation analysis revealed a correlation between differentially metabolites and the gut microbiota. Lactobacillus and Akkermansia are the core genera. CONCLUSION RGS can be a candidate for the prevention and amelioration of hyperuricemia and concomitant sperm damage. Its mechanism of action is closely related to sphingolipid metabolism, pyrimidine metabolism, and the modulation of gut microbiota, such as Lactobacillus and Akkermansia.
Collapse
Affiliation(s)
- Xueying Ji
- Clinical Nutrition Department, Weifang People's Hospital, Shandong Second Medical University, Weifang, 261041, Shandong, China; School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Lingbo Yu
- Clinical Nutrition Department, Weifang People's Hospital, Shandong Second Medical University, Weifang, 261041, Shandong, China
| | - Chengcheng Han
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Hui Gao
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Yuqing Cai
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Jiamin Li
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Yi He
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Hao Lu
- School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Guihua Song
- Clinical Nutrition Department, Weifang People's Hospital, Shandong Second Medical University, Weifang, 261041, Shandong, China.
| | - Peng Xue
- Clinical Nutrition Department, Weifang People's Hospital, Shandong Second Medical University, Weifang, 261041, Shandong, China; School of Public Health, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
4
|
Chen H, Dong M, He H, Piao X, Han X, Li R, Jiang H, Li X, Li B, Cui R. Ginsenoside Re Prevents Depression-like Behaviors via Inhibition of Inflammation, Oxidative Stress, and Activating BDNF/TrkB/ERK/CREB Signaling: An In Vivo and In Vitro Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19838-19851. [PMID: 39186472 DOI: 10.1021/acs.jafc.4c04394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Depression is a widespread disease, with high mortality and recurrence rates. Recent studies have shown that elevated cytokine levels are implicated in the molecular mechanisms of depression. Oxidative stress contributes to the stimulation of cytokine production. Growing evidence suggests that ginsenoside Re (Gs-Re) exerts a neuroprotective effect on the hippocampus by suppressing oxidative stress and inflammation. However, the effect and mechanism of Gs-Re in the treatment of depression remain understudied. This study aimed to evaluate the neuroprotective and antidepressant-like effects of Gs-Re and the possible underlying mechanisms. In this article, the antidepressant-like effect of the Gs-Re was studied both in vitro (H2O2-induced oxidative stress in HT-22 cells) and in vivo (reserpine-induced depressive model mice). Our results indicated that, at the cellular level, Gs-Re effectively enhanced cell survival following H2O2 stimulation, inhibited the mass production of oxidative stress markers (MDA and ROS), and prevented the occurrence of apoptosis. Moreover, Gs-Re significantly reduced the levels of proinflammatory cytokines IL-1β, IL-6, and TNF-α and restored the abnormal mitochondrial membrane potential. Subsequently, Gs-Re treatment reversed reserpine-induced neuroinflammation and depressive-like behaviors in vivo and inhibited microglia overactivation. Furthermore, the alterations in the BDNF/TrkB/ERK/CREB signaling pathway induced by H2O2 or reserpine in HT-22 cells or in the mouse hippocampus were significantly reversed by Gs-Re. K252a blocked the improvement of Gs-Re on depression-like behavior and eliminated the inhibition of oxidative stress and neuroinflammation in vivo. This study suggested that Gs-Re produces neuroprotective and depressive effects by inhibiting oxidative stress and inflammation and activating the BDNF/TrkB/ERK/CREB pathway.
Collapse
Affiliation(s)
- Hongyu Chen
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Mengmeng Dong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Huihan He
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Xinmiao Piao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Xu Han
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Runxin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Huiyi Jiang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Xin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Jilin Engineering Laboratory for Screening of Antidepressants, Changchun 130041, P. R. China
| |
Collapse
|
5
|
Han L, Chen W, Li J, Zhao Y, Zong Y, He Z, Du R. Palmatine improves cognitive dysfunction in Alzheimer's disease model rats through autophagy pathway and regulation of gut microbiota. Brain Res 2024; 1835:148932. [PMID: 38609032 DOI: 10.1016/j.brainres.2024.148932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/17/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Alzheimer's disease (AD) is a primary degenerative encephalopathy that first appeared as a decline in memory and learning skills. Over time, the condition's severity grew. Palmatine (Pal) alleviates Alzheimer's disease symptoms, which has neuroprotective benefits. Numerous investigations have demonstrated a close relationship among AD and gut structure changes. The aim of the research was investigating whether the improvement of Pal on AD is linked to regulating gut flora and autophagy. First, we used Aβ1-40 to induce apoptosis in HT22 cells. After Pal treatment, apoptosis can be improved. Then, We used bilateral intracranial hippocampal injection of Aβ1-40 for establishing the AD model, after treatment with Pal, the morris water maze experiment and eight-arm maze test demonstrated that Pal enhanced the AD rats' capacity for learning and memory, HE staining illustrated that Pal improved the morphological abnormalities of brain cells and gut tissue damage. Pal reduced the death of hippocampus neurons, as shown by Nissl staining. Pal substantially reduced Tau hyperphosphorylation and Aβ accumulation in the brain, according to immunohistochemical labelling. Pal improved the expression of LC3, Beclin 1, AMPK, and suppressed the expression of mTOR and P62, as validated by RT-qPCR and immunofluorescence labelling. This suggests that Pal's treatment of AD may be associated with the control of the AMPK/mTOR autophagy signalling system. 16S rRNA sequencing and short-chain fatty acids (SCFAs) content detection analysis illustrated that Pal has the potential to enhance the content of SCFAs, reverse the alterations in gut microorganisms. It has been showed by the study that Pal could improve AD by activating autophagy signaling pathway and improving gut barrier changes.
Collapse
Affiliation(s)
- Lu Han
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Weijia Chen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jianming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Ying Zong
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Zhongmei He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China.
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education of China, Changchun 130118, China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer of China, Changchun 130118, China.
| |
Collapse
|
6
|
Urquiza-Martínez MV, Fabián-Avilés IM, Torner L, Servín-Campuzano H, González-Avilés M. Integrative Approach of Treating Early Undernutrition with an Enriched Black Corn Chip, Study on a Murine Model. Nutrients 2024; 16:2001. [PMID: 38999749 PMCID: PMC11243394 DOI: 10.3390/nu16132001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 07/14/2024] Open
Abstract
Undernutrition (UN) increases child vulnerability to illness and mortality. Caused by a low amount and/or poor quality of food intake, it impacts physical, cognitive, and social development. Modern types of food consumption have given highly processed food a higher cultural value compared to minimally processed food. OBJECTIVE The objective of this study was to evaluate the effect on growth, metabolism, physical activity (PA), memory, inflammation, and toxicity of an enriched black corn chip (BC) made with endemic ingredients on post-weaned UN mice. METHODS A chip was made with a mixture of black corn, fava beans, amaranth, and nopal cactus. To probe the effects of UN, UN was induced in 3wo post-weaned male C57Bl/6j mice through a low-protein diet (LPD-50% of the regular requirement of protein) for 3w. Then, the BC was introduced to the animals' diet (17%) for 5w; murinometric parameters were measured, as were postprandial glucose response, PA, and short-term memory. Histological analysis was conducted on the liver and kidneys to measure toxicity. Gene expression related to energy balance, thermogenesis, and inflammation was measured in adipose and hypothalamic tissues. RESULTS Treatment with the BC significantly improved mouse growth, even with a low protein intake, as evidenced by a significant increase in body weight, tail length, cerebral growth, memory improvement, physical activation, normalized energy expenditure (thermogenesis), and orexigenic peptides (AGRP and NPY). It decreased anorexigenic peptides (POMC), and there was no tissue toxicity. CONCLUSIONS BC treatment, even with persistent low protein intake, is a promising strategy against UN, as it showed efficacy in correcting growth deficiency, cognitive impairment, and metabolic problems linked to treatment by adjusting energy expenditure, which led to the promotion of energy intake and regulation of thermogenesis, all by using low-cost, accessible, and endemic ingredients.
Collapse
Affiliation(s)
- Mercedes-Victoria Urquiza-Martínez
- Master in Engineering for the Energetic Sustainability, Universidad Intercultural Indígena de Michoacán, Campus Tzipekua, Pátzcuaro 61614, Mexico
| | - Imelda M Fabián-Avilés
- Master in Engineering for the Energetic Sustainability, Universidad Intercultural Indígena de Michoacán, Campus Tzipekua, Pátzcuaro 61614, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia 58330, Mexico
| | - Hermelinda Servín-Campuzano
- Master in Engineering for the Energetic Sustainability, Universidad Intercultural Indígena de Michoacán, Campus Tzipekua, Pátzcuaro 61614, Mexico
| | - Mauricio González-Avilés
- Master in Engineering for the Energetic Sustainability, Universidad Intercultural Indígena de Michoacán, Campus Tzipekua, Pátzcuaro 61614, Mexico
| |
Collapse
|
7
|
Li K, Li J, Li Z, Men L, Zuo H, Gong X. Cisplatin-based combination therapies: Their efficacy with a focus on ginsenosides co-administration. Pharmacol Res 2024; 203:107175. [PMID: 38582357 DOI: 10.1016/j.phrs.2024.107175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Cisplatin, a frequently prescribed chemotherapeutic agent, serves as a clinically therapeutic strategy for a broad range of malignancies. Its primary mode of action centers around interference with DNA replication and RNA transcription, thereby inducing apoptosis in cancer cells. Nevertheless, the clinical utility of cisplatin is constrained by its severe adverse effects and the burgeoning problem of drug resistance. Ginsenosides, potent bioactive constituents derived from ginseng, possess an array of biological activities. Recent scientific investigations underscore the substantial amplification of cisplatin's anticancer potency and the mitigation of its harmful side effects when administered concomitantly with ginsenosides. This review aims to explore the underlying mechanisms at play in this combination therapy. Initially, we provide a concise introduction to the cisplatin. Then, we pivot towards illuminating how ginsenosides bolster the anticancer efficacy of cisplatin and counteract cisplatin resistance, culminating in enhanced therapeutic outcomes. Furthermore, we provide an extensive discussion on the reduction of cisplatin-induced toxicity in the kidneys, liver, gastrointestinal tract, nervous system, and ear, accompanied by immune-fortification with ginsenosides. The existing clinical combined use of cisplatin and ginsenosides is also discussed. We propose several recommendations to propel additional research into the mechanisms governing the synergistic use of ginsenosides and cisplatin, thereby furnishing invaluable insights and fostering advancement in combined modality therapy.
Collapse
Affiliation(s)
- Keke Li
- Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Jiwen Li
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China
| | - Zhongyu Li
- Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Lei Men
- Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Haibin Zuo
- School of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Xiaojie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, Ministry of Education, Dalian Minzu University, Dalian 116600, China; School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
8
|
Zhang J, Ma F, Li Z, Li Y, Sun X, Song M, Yang F, Wu E, Wei X, Wang Z, Yang L. NFKB2 mediates colorectal cancer cell immune escape and metastasis in a STAT2/PD‐L1‐dependent manner. MedComm (Beijing) 2024; 5:e521. [PMID: 38660687 PMCID: PMC11042535 DOI: 10.1002/mco2.521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 04/26/2024] Open
Abstract
This study systematically analyzed the molecular mechanism and function of nuclear factor kappa B subunit 2 (NFKB2) in colorectal cancer (CRC) to investigate the potential of NFKB2 as a therapeutic target for CRC. Various experimental techniques, including RNA sequencing, proteome chip assays, and small molecule analysis, were used to obtain a deeper understanding of the regulation of NFKB2 in CRC. The results revealed that NFKB2 was upregulated in a significant proportion of patients with advanced hepatic metastasis of CRC. NFKB2 played an important role in promoting tumor growth through CD8+ T-cell exhaustion. Moreover, NFKB2 directly interacted with signal transducer and activator of transcription 2 (STAT2), leading to increased phosphorylation of STAT2 and the upregulation of programmed death ligand 1 (PD-L1). Applying a small molecule inhibitor of NFKB2 (Rg5) led to a reduction in PD-L1 expression and improved response to programmed death-1 blockade-based immunotherapy. In conclusion, the facilitated NFKB2-STAT2/PD-L1 axis may suppress immune surveillance in CRC and targeting NFKB2 may enhance the efficacy of immunotherapeutic strategies. Our results provide novel insights into the molecular mechanisms underlying the contribution of NFKB2 in CRC immune escape.
Collapse
Affiliation(s)
- Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Fen Ma
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhe Li
- Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yuan Li
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xun Sun
- Gastrointestinal SurgeryLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mingxu Song
- Human Reproductive and Genetic CenterAffiliated Hospital of Jiangnan UniversityJiangsuChina
| | - Fan Yang
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Enjiang Wu
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiaohui Wei
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhengtao Wang
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Li Yang
- Shanghai Key Laboratory of Compound Chinese MedicinesThe MOE Key Laboratory for Standardization of Chinese MedicinesInstitute of Chinese Materia MedicaShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
9
|
Luo Y, Zhang J, Jiao Y, Huang H, Ming L, Song Y, Niu Y, Tang X, Liu L, Li Y, Jiang Y. Dihydroartemisinin abolishes cisplatin-induced nephrotoxicity in vivo. J Nat Med 2024; 78:439-454. [PMID: 38351420 DOI: 10.1007/s11418-024-01783-5] [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: 09/30/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024]
Abstract
Dihydroartemisinin (DHA), a derivative of artemisinin which is primarily used to treat malaria in clinic, also confers protective effect on lipopolysaccharide-induced nephrotoxicity. While, the activities of DHA in cisplatin (CDDP)-caused nephrotoxicity are elusive. To investigate the role and underlying mechanism of DHA in CDDP-induced nephrotoxicity. Mice were randomly separated into four groups: normal, CDDP, and DHA (25 and 50 mg/kg were orally injected 1 h before CDDP for consecutive 10 days). All mice except the normal were single injected intraperitoneally with CDDP (22 mg/kg) for once on the 7th day. Combined with quantitative proteomics and bioinformatics analysis, the impact of DHA on renal cell apoptosis, oxidative stress, biochemical indexes, and inflammation in mice were investigated. Moreover, a human hepatocellular carcinoma cells xenograft model was established to elucidate the impact of DHA on tumor-related effects of CDDP. DHA reduced the levels of creatinine (CREA) (p < 0.01) and blood urea nitrogen (BUN) (p < 0.01), reversed CDDP-induced oxidative, inflammatory, and apoptosis indexes (p < 0.01). Mechanistically, DHA attenuated CDDP-induced inflammation by inhibiting nuclear factor κB p65 (NFκB p65) expression, and suppressed CDDP-induced renal cell apoptosis by inhibiting p63-mediated endogenous and exogenous apoptosis pathways. Additionally, DHA alone significantly decreased the tumor weight and did not destroy the antitumor effect of CDDP, and did not impact AST and ALT. In conclusion, DHA prevents CDDP-triggered nephrotoxicity via reducing inflammation, oxidative stress, and apoptosis. The mechanisms refer to inhibiting NFκB p65-regulated inflammation and alleviating p63-mediated mitochondrial endogenous and Fas death receptor exogenous apoptosis pathway.
Collapse
Affiliation(s)
- Yan Luo
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Jiaxing Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yue Jiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research On Prevention and Treatment of Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Liangshan Ming
- Institute for Advanced Study, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Yunlei Song
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Yanlong Niu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Xiaolu Tang
- Department of Human Anatomy, School of Basic Medical Science, Gannan Medical University, Jiangxi, China
| | - Liwei Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Yumao Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China.
- Artemisinin Research Center, and Institute of Chinese Materia Medical, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
10
|
Ahmad SS, Ahmad K, Hwang YC, Lee EJ, Choi I. Therapeutic Applications of Ginseng Natural Compounds for Health Management. Int J Mol Sci 2023; 24:17290. [PMID: 38139116 PMCID: PMC10744087 DOI: 10.3390/ijms242417290] [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: 11/15/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Ginseng is usually consumed as a daily food supplement to improve health and has been shown to benefit skeletal muscle, improve glucose metabolism, and ameliorate muscle-wasting conditions, cardiovascular diseases, stroke, and the effects of aging and cancers. Ginseng has also been reported to help maintain bone strength and liver (digestion, metabolism, detoxification, and protein synthesis) and kidney functions. In addition, ginseng is often used to treat age-associated neurodegenerative disorders, and ginseng and ginseng-derived natural products are popular natural remedies for diseases such as diabetes, obesity, oxidative stress, and inflammation, as well as fungal, bacterial, and viral infections. Ginseng is a well-known herbal medication, known to alleviate the actions of several cytokines. The article concludes with future directions and significant application of ginseng compounds for researchers in understanding the promising role of ginseng in the treatment of several diseases. Overall, this study was undertaken to highlight the broad-spectrum therapeutic applications of ginseng compounds for health management.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (S.S.A.); (K.A.); (Y.C.H.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (S.S.A.); (K.A.); (Y.C.H.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Ye Chan Hwang
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (S.S.A.); (K.A.); (Y.C.H.); (E.J.L.)
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (S.S.A.); (K.A.); (Y.C.H.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (S.S.A.); (K.A.); (Y.C.H.); (E.J.L.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
11
|
Zhang D, Luo G, Jin K, Bao X, Huang L, Ke J. The underlying mechanisms of cisplatin-induced nephrotoxicity and its therapeutic intervention using natural compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2925-2941. [PMID: 37289283 DOI: 10.1007/s00210-023-02559-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/28/2023] [Indexed: 06/09/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug widely used for the treatment of various solid tumors; however, its clinical use and efficacy are limited by its inherent nephrotoxicity. The pathogenesis of cisplatin-induced nephrotoxicity is complex and has not been fully elucidated. Cellular uptake and transport, DNA damage, apoptosis, oxidative stress, inflammatory response, and autophagy are involved in the development of cisplatin-induced nephrotoxicity. Currently, despite some deficiencies, hydration regimens remain the major protective measures against cisplatin-induced nephrotoxicity. Therefore, effective drugs must be explored and developed to prevent and treat cisplatin-induced kidney injury. In recent years, many natural compounds with high efficiency and low toxicity have been identified for the treatment of cisplatin-induced nephrotoxicity, including quercetin, saikosaponin D, berberine, resveratrol, and curcumin. These natural agents have multiple targets, multiple effects, and low drug resistance; therefore, they can be safely used as a supplementary regimen or combination therapy for cisplatin-induced nephrotoxicity. This review aimed to comprehensively describe the molecular mechanisms underlying cisplatin-induced nephrotoxicity and summarize natural kidney-protecting compounds to provide new ideas for the development of better therapeutic agents.
Collapse
Affiliation(s)
- Doudou Zhang
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Guangwen Luo
- Jinhua Municipal Central Hospital, Jinhua, 321000, China.
| | - Kaixiang Jin
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Xiaodong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lili Huang
- Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, China
| | - Jianghuan Ke
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| |
Collapse
|
12
|
Hou YY, Qi SM, Leng J, Shen Q, Tang S, Zhang JT, Hu JN, Jiang S, Li W. Lobetyolin, a Q-marker isolated from Radix Platycodi, exerts protective effects on cisplatin-induced cytotoxicity in HEK293 cells. J Nat Med 2023; 77:721-734. [PMID: 37353674 DOI: 10.1007/s11418-023-01714-w] [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: 10/12/2022] [Accepted: 05/28/2023] [Indexed: 06/25/2023]
Abstract
This study investigated the protective effect of lobetyolin (LBT), a Q-marker isolated from the roots of Platycodon grandiflorum (Radix Platycodi), against cisplatin-induced cytotoxicity in human embryonic kidney (HEK293) cells. Results showed that LBT at 20 μM significantly prevented cisplatin-induced cytotoxicity by improving the viability of HEK293 cells, decreasing levels of MDA, and decreasing GSH content triggered by cisplatin. It also suppressed reactive oxygen species (ROS) levels. Molecular docking analysis revealed a strong binding affinity between LBT and the NF-κB protein, with a docking fraction of - 6.5 kcal/mol. These results provide compelling evidence suggesting a potential link between the visualization analysis of LBT and its protective mechanism, specifically implicating the NF-κB signaling pathway. LBT also reduced the expression level of tumor necrosis factor-alpha (TNF-α), phosphorylation NF-κB and IκBα in HEK293 cells which were increased by cisplatin exposure, leading to inhibition of inflammation. Furthermore, western blotting showed that LBT antagonized the up-regulation of Bax, cleaved caspase 3, 8, and 9 expression and inhibited the MAPK signaling pathway by down-regulating phosphorylation JNK, ERK, and p38, partially ameliorating cisplatin-induced cytotoxicity in HEK293 cells. Therefore, these results indicate that LBT has potentially protected renal function by inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Yun-Yi Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Si-Min Qi
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Leng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Qiong Shen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shan Tang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
- National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| |
Collapse
|
13
|
Luo X, Xie D, Chen Z, Ji Q. Protective effects of ginsenosides in cisplatin-induced kidney injury: A systematic review, meta-analysis. Indian J Pharmacol 2023; 55:243-250. [PMID: 37737077 PMCID: PMC10657623 DOI: 10.4103/ijp.ijp_251_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/23/2023] Open
Abstract
Although evidence suggests ginsenosides, the primary active and distinctive components of ginseng, have beneficial effects in cisplatin-induced nephrotoxicity, their efficacy and protective mechanisms remain unclear. The aim of the current meta-analysis is to study the effectiveness and mechanisms of ginsenosides in a model of nephrotoxicity induced by cisplatin. Preclinical investigations were conducted in the search of various databases including Medline, Web of Science, Google, CNKI, Embase, and the Wanfang database. 12 studies with 216 animals were included in this review. Stata 15.0 and RevMan 5.3 were used for statistical analyses. The pooled results showed that ginsenosides significantly improved kidney function, and inhibited histological damage. The protective mechanism of ginsenosides is associated with its antioxidative stress, anti-inflammation, anti-apoptosis, and anti-autophagy. The results of our study indicate that ginsenosides have the potential to mitigate nephrotoxicity induced by cisplatin through the modulation of various targets and pathways. Consequently, ginsenosides hold promise as therapeutic agents for the clinical management and prevention of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Xinyi Luo
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Dengpiao Xie
- Department of Kidney, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ziwei Chen
- Department of Kidney, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Qing Ji
- Department of Kidney, Chengdu First People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Chen J, Li M, Huo X, Li Z, Qu D, Sha J, Sun Y. A Novel Process for One-Step Separation and Cyclodextrin Inclusion of Ginsenoside Rg5 from Ginseng Stem-Leaf Saponins (GSLS): Preparation, Characterization, and Evaluation of Storage Stability and Bioactivity. Foods 2023; 12:2349. [PMID: 37372560 DOI: 10.3390/foods12122349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Ginsenoside Rg5 has been proven to possess numerous health benefits. However, Rg5 is difficult to prepare using the current methods, and the poor stability and solubility of Rg5 are intractable properties that limit its application. We try to establish and optimize a new method for preparing Rg5. METHODS Different amino acids acted as catalysts, and reaction conditions were investigated to transform Rg5 in GSLS. Different CDs and reaction conditions were investigated for the preparation of CD-Rg5 based on yield and purity; ESI-MS, FT-IR, XRD and SEM analyses were used to prove the formation of the CD-Rg5 inclusion complex. Both the stability and bioactivity of β-CD-Rg5 were investigated. RESULTS The content of Rg5 reached 140.8 mg/g after transformation of GSLS using Asp as a catalyst. The yield of β-CD-Rg5 reached a maximum of 12% and a purity of 92.5%. The results showed that the β-CD-Rg5 inclusion complex can improve its stability of Rg5 against light and temperature. Antioxidant activity analyses against DPPH, ABTS+, and Fe2+ chelation showed enhanced antioxidant activity of the β-CD-Rg5 inclusion complex. CONCLUSIONS A novel and effective strategy for the separation of Rg5 from ginseng stem-leaf saponins (GSLS) was developed to improve the stability, solubility, and bioactivity of Rg5.
Collapse
Affiliation(s)
- Jianbo Chen
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Meijia Li
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Xiaohui Huo
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Zhiman Li
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Di Qu
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Jiyue Sha
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| | - Yinshi Sun
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agriculture Sciences, Changchun 130112, China
| |
Collapse
|
15
|
Li J, Li T, Li Z, Song Z, Gong X. Potential therapeutic effects of Chinese meteria medica in mitigating drug-induced acute kidney injury. Front Pharmacol 2023; 14:1153297. [PMID: 37077810 PMCID: PMC10106589 DOI: 10.3389/fphar.2023.1153297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Drug-induced acute kidney injury (DI-AKI) is one of the leading causes of kidney injury, is associated with high mortality and morbidity, and limits the clinical use of certain therapeutic or diagnostic agents, such as antineoplastic drugs, antibiotics, immunosuppressants, non-steroidal anti-inflammatory drugs, and contrast media. In recent years, numerous studies have shown that many Chinese meteria medica, metabolites derived from botanical drugs, and Chinese medicinal formulas confer protective effects against DI-AKI by targeting a variety of cellular or molecular mechanisms, such as oxidative stress, inflammatory, cell necrosis, apoptosis, and autophagy. This review summarizes the research status of common DI-AKI with Chinese meteria medica interventions, including cisplatin, gentamicin, contrast agents, methotrexate, and acetaminophen. At the same time, this review introduces the metabolites with application prospects represented by ginseng saponins, tetramethylpyrazine, panax notoginseng saponins, and curcumin. Overall, this review provides a reference for the development of promising nephroprotectants.
Collapse
|
16
|
Ghosh A, Banik S, Suzuki Y, Mibe Y, Rikimura S, Komamoto T, Kuromi K, Yamada K, Sato H, Onoue S. Lysophosphatidylcholine-based liposome to improve oral absorption and nephroprotective effects of astaxanthin. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:2981-2988. [PMID: 36350072 DOI: 10.1002/jsfa.12329] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The present study was aimed to develop astaxanthin (AX)-loaded liposomes by the utilization of soybean phosphatidylcholine (PC) and lysophosphatidylcholine (LPC) to improve the nutraceutical properties of AX. AX-loaded liposomes consisting of PC (PC/AX) and LPC (LPC/AX) were evaluated in terms of particle size distribution, morphology, release characteristics, pharmacokinetic behavior, and nephroprotective effects in a rat model of acute kidney injury. RESULTS PC/AX and LPC/AX had uniform size distributions with a mean particle size of 254 and 148 nm, respectively. Under pH 6.8 conditions, both liposomes exhibited improved dissolution behavior of AX compared with crystalline AX (cAX). In particular, LPC/AX showed a sevenfold higher release of AX than PC/AX. After the oral administration of LPC/AX (33.2 mg AX kg-1 ) to rats, there was a significant increase in systemic exposure to AX, as evidenced by a 15-fold higher AUC0-24 h than PC/AX. However, the oral absorption of AX in the cAX group was negligible. Based on the results of histological analysis and measurement of plasma biomarkers, LPC/AX exhibited improved nephroprotective effects of AX in the rat model of kidney injury. CONCLUSION From these observations, a strategic application of the LPC-based liposomal approach might be a promising option to improve the nutraceutical properties of AX. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Antara Ghosh
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Sujan Banik
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yui Suzuki
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | | | | | | | - Kohei Yamada
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hideyuki Sato
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Satomi Onoue
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
17
|
Lu S, Li Y, Qian Z, Zhao T, Feng Z, Weng X, Yu L. Role of the inflammasome in insulin resistance and type 2 diabetes mellitus. Front Immunol 2023; 14:1052756. [PMID: 36993972 PMCID: PMC10040598 DOI: 10.3389/fimmu.2023.1052756] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
The inflammasome is a protein complex composed of a variety of proteins in cells and which participates in the innate immune response of the body. It can be activated by upstream signal regulation and plays an important role in pyroptosis, apoptosis, inflammation, tumor regulation, etc. In recent years, the number of metabolic syndrome patients with insulin resistance (IR) has increased year by year, and the inflammasome is closely related to the occurrence and development of metabolic diseases. The inflammasome can directly or indirectly affect conduction of the insulin signaling pathway, involvement the occurrence of IR and type 2 diabetes mellitus (T2DM). Moreover, various therapeutic agents also work through the inflammasome to treat with diabetes. This review focuses on the role of inflammasome on IR and T2DM, pointing out the association and utility value. Briefly, we have discussed the main inflammasomes, including NLRP1, NLRP3, NLRC4, NLRP6 and AIM2, as well as their structure, activation and regulation in IR were described in detail. Finally, we discussed the current therapeutic options-associated with inflammasome for the treatment of T2DM. Specially, the NLRP3-related therapeutic agents and options are widely developed. In summary, this article reviews the role of and research progress on the inflammasome in IR and T2DM.
Collapse
Affiliation(s)
- Shen Lu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanrong Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhaojun Qian
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tiesuo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaogang Weng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| | - Lili Yu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, China
- *Correspondence: Lili Yu, ; Xiaogang Weng,
| |
Collapse
|
18
|
Oral hydrogel microspheres were used for highly specific delivery of Steamed Codonopsis lanceolata to exert the protective effects on cisplatin-induced acute kidney injury in mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
19
|
Fan M, Lan X, Wang Q, Shan M, Fang X, Zhang Y, Wu D, Luo H, Gao W, Zhu D. Renal function protection and the mechanism of ginsenosides: Current progress and future perspectives. Front Pharmacol 2023; 14:1070738. [PMID: 36814491 PMCID: PMC9939702 DOI: 10.3389/fphar.2023.1070738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
Nephropathy is a general term for kidney diseases, which refers to changes in the structure and function of the kidney caused by various factors, resulting in pathological damage to the kidney, abnormal blood or urine components, and other diseases. The main manifestations of kidney disease include hematuria, albuminuria, edema, hypertension, anemia, lower back pain, oliguria, and other symptoms. Early detection, diagnosis, and active treatment are required to prevent chronic renal failure. The concept of nephropathy encompasses a wide range of conditions, including acute renal injury, chronic kidney disease, nephritis, renal fibrosis, and diabetic nephropathy. Some of these kidney-related diseases are interrelated and may lead to serious complications without effective control. In serious cases, it can also develop into chronic renal dysfunction and eventually end-stage renal disease. As a result, it seriously affects the quality of life of patients and places a great economic burden on society and families. Ginsenoside is one of the main active components of ginseng, with anti-inflammatory, anti-tumor, antioxidant, and other pharmacological activities. A variety of monomers in ginsenosides can play protective roles in multiple organs. According to the difference of core structure, ginsenosides can be divided into protopanaxadiol-type (including Rb1, Rb3, Rg3, Rh2, Rd and CK, etc.), and protopanaxatriol (protopanaxatriol)- type (including Rg1, Rg2 and Rh1, etc.), and other types (including Rg5, Rh4, Rh3, Rk1, and Rk3, etc.). All of these ginsenosides showed significant renal function protection, which can reduce renal damage in renal injury, nephritis, renal fibrosis, and diabetic nephropathy models. This review summarizes reports on renal function protection and the mechanisms of action of these ginsenosides in various renal injury models.
Collapse
Affiliation(s)
- Meiling Fan
- The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xintian Lan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Qunling Wang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Mengyao Shan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Xiaoxue Fang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Yegang Zhang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Donglu Wu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China,School of Clinical Medical, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Wenyi Gao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,*Correspondence: Wenyi Gao, ; Difu Zhu,
| | - Difu Zhu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China,*Correspondence: Wenyi Gao, ; Difu Zhu,
| |
Collapse
|
20
|
Heo H, Kim Y, Cha B, Brito S, Kim H, Kim H, Fatombi BM, Jung SY, Lee SM, Lei L, Lee SH, Park GW, Kwak BM, Bin BH, Park JH, Lee MG. A systematic exploration of ginsenoside Rg5 reveals anti-inflammatory functions in airway mucosa cells. J Ginseng Res 2023; 47:97-105. [PMID: 36644392 PMCID: PMC9834007 DOI: 10.1016/j.jgr.2022.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
Background Hyperactivated airway mucosa cells overproduce mucin and cause severe breathing complications. Here, we aimed to identify the effects of saponins derived from Panax ginseng on inflammation and mucin overproduction. Methods NCI-H292 cells were pre-incubated with 16 saponins derived from P. ginseng, and mucin overproduction was induced by treatment with phorbol 12-myristate 13-acetate (PMA). Mucin protein MUC5AC was quantified by enzyme-linked immunosorbent assay, and mRNA levels were analyzed using quantitative polymerase chain reaction (qPCR). Moreover, we performed a transcriptome analysis of PMA-treated NCI-H292 cells in the absence or presence of Rg5, and differential gene expression was confirmed using qPCR. Phosphorylation levels of signaling molecules, and the abundance of lipid droplets, were measured by western blotting, flow cytometry, and confocal microscopy. Results Ginsenoside Rg5 effectively reduced MUC5AC secretion and decreased MUC5AC mRNA levels. A systematic functional network analysis revealed that Rg5 upregulated cholesterol and glycerolipid metabolism, resulting in the production of lipid droplets to clear reactive oxygen species (ROS), and modulated the mitogen-activated protein kinase and nuclear factor (NF)-κB signaling pathways to regulate inflammatory responses. Rg5 induced the accumulation of lipid droplets and decreased cellular ROS levels, and N-acetyl-l-cysteine, a ROS inhibitor, reduced MUC5AC secretion via Rg5. Furthermore, Rg5 hampered the phosphorylation of extracellular signal-regulated kinase and p38 proteins, affecting the NF-κB signaling pathway and pro-inflammatory responses. Conclusion Rg5 alleviated inflammatory responses by reducing mucin secretion and promoting lipid droplet-mediated ROS clearance. Therefore, Rg5 may have potential as a therapeutic agent to alleviate respiratory disorders caused by hyperactivation of mucosa cells.
Collapse
Affiliation(s)
- Hyojin Heo
- Department of Applied Biotechnology, Ajou University, Suwon, Republic of Korea
| | - Yumin Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea,Department of Biomedical Science and Engineering, Gwangju Institute of Science & Technology, Republic of Korea
| | - Byungsun Cha
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Sofia Brito
- Department of Applied Biotechnology, Ajou University, Suwon, Republic of Korea
| | - Haneul Kim
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Hyunjin Kim
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | | | - So Young Jung
- Department of Applied Biotechnology, Ajou University, Suwon, Republic of Korea
| | - So Min Lee
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Lei Lei
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Sang Hun Lee
- Department of Biological Sciences, Ajou University, Suwon, Republic of Korea
| | - Geon-woo Park
- Department of Applied Biotechnology, Ajou University, Suwon, Republic of Korea,Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea
| | - Byeong-Mun Kwak
- Department of Meridian and Acupoint, College of Korean Medicine, Semyung University, Chungbuk, Republic of Korea,Corresponding author. Department of Meridian and Acupoint, College of Korean Medicine, Semyung University, Chungbuk, 27136, Republic of Korea.
| | - Bum-Ho Bin
- Department of Applied Biotechnology, Ajou University, Suwon, Republic of Korea,Department of Biological Sciences, Ajou University, Suwon, Republic of Korea,Corresponding author. Department of Biological Sciences, Ajou University, Suwon, 16499, Republic of Korea.
| | - Ji-Hwan Park
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea,Corresponding author. Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, Republic of Korea.
| | - Mi-Gi Lee
- Bio-Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea,Corresponding author. Bio-Center, Gyeonggi-do Business and Science Accelerator, Suwon, 16229, Republic of Korea.
| |
Collapse
|
21
|
Zhang H, Liu W, Qi SM, Chi JF, Gao Q, Lin XH, Ren S, Wang Z, Lei XJ, Li W. Improved effect of fresh ginseng paste (radix ginseng-ziziphus jujube) on hyperuricemia based on network pharmacology and molecular docking. Front Pharmacol 2022; 13:955219. [PMID: 36386218 PMCID: PMC9641371 DOI: 10.3389/fphar.2022.955219] [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: 05/28/2022] [Accepted: 10/10/2022] [Indexed: 08/02/2024] Open
Abstract
Background: Hyperuricemia (HUA) is a metabolic disease caused by reduced excretion or increased production of uric acid. This research aims to study the practical components, active targets, and potential mechanism of the "Radix ginseng (RG)-Ziziphus jujube (ZJ)" herb pair through molecular docking, network pharmacology, and animal experiments. Methods: The potential targets of "Radix ginseng (RG)-Ziziphus jujube (ZJ)" herb pair were obtained from the TCMSP database. The therapeutic targets of HUA were acquired from the GendCards, OMIM, PharmGkb, and TTD databases. Protein-protein interaction network (PPI) was constructed in the STRING 11.0 database. The David database was used for enrichment analysis. Molecular Docking was finished by the AutoDock Vina. And we employed Radix ginseng and Ziziphus jujube as raw materials, which would develop a new functional food fresh ginseng paste (FGP) after boiling. In addition, benzbromarone (Ben) (7.8 mg/kg) and allopurinol (All) (5 mg/kg) were used as positive drugs to evaluate the hyperuricemia induced by FGP (400 and 800 mg/kg) potassium oxazine (PO) (100 mg/kg) and hypoxanthine (HX) (500 mg/kg) on mice. Results: The results showed that 25 targets in the "RG-ZJ" herb pair interacted with hyperuricemia. Then protein-protein interaction (PPI) analysis showed that TNF, IL-1β, and VEGFA were core genes. KEGG enrichment analysis showed that the Toll-like receptor signaling pathway and IL-17 signaling pathway were mainly involved. Meantime, animal experiments showed that FGP could improve the HUA status of mice by reducing serum UA BUN, XO, and liver XO levels (p < 0.05, p < 0.01). Furthermore, we analyzed the main ingredients of FGP by HPLC. We found that the main ingredients of FGP had solid binding activity to the core target of HUA by molecular docking. Conclusion: This study explored the active ingredients and targets of the "RG-ZJ" herb pair on HUA through network pharmacology, molecular docking, and animal experiments. It revealed the improvement of FGP in mice with HUA.
Collapse
Affiliation(s)
- Hao Zhang
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Wei Liu
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Si-Min Qi
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Jian-Feng Chi
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Qiang Gao
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Xiang-Hui Lin
- Liaoning Xifeng Pharmaceutical Group Co., Ltd., Huanren, China
| | - Shen Ren
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Zi Wang
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Xiu-juan Lei
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| |
Collapse
|
22
|
Liu YB, Sun DZ, Chen KC, Zhang JJ, Hou YY, Gao XF, Cai EB, Zhu HY, Zheng YN, Chen RX, Liu S, Li W. Based on molecular docking to evaluate the protective effect of saponins from ginseng berry on D-gal-induced brain injury via multiple molecular mechanisms in mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
23
|
Wang X, Wang R, Qiao Y, Li Y. Progress on the efficacy and mechanism of action of panax ginseng monomer saponins treat toxicity. Front Pharmacol 2022; 13:1022266. [PMID: 36199681 PMCID: PMC9527293 DOI: 10.3389/fphar.2022.1022266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/05/2022] [Indexed: 12/06/2022] Open
Abstract
As a traditional Chinese herbal medicine, Panax ginseng C. A. Meyer (PG) has preventive and therapeutic effects on various diseases. Ginsenosides are main active ingredients of PG and have good pharmacological effects. Due to the diversity of chemical structures and physicochemical properties of ginsenosides, Currently, related studies on PG monomer saponins are mainly focused on the cardiovascular system, nervous system, antidiabetic, and antitumor. There are few types of research on the toxin treatment, predominantly exogenous toxicity. PG and its monomer ginsenosides are undoubtedly a practical option for treating exogenous toxicity for drug-induced or metal-induced side effects such as nephrotoxicity, hepatotoxicity, cardiotoxicity, metal toxicity and other exogenous toxicity caused by drugs or metals. The mechanism focuses on antioxidant, anti-inflammatory, and anti-apoptotic, as well as modulation of signaling pathways. It summarized the therapeutic effects of ginseng monomer saponins on exogenous toxicity and demonstrated that ginsenosides could be used as potential drugs to treat exogenous toxicity and reduce drug toxicities.
Collapse
Affiliation(s)
- Xinyi Wang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Rongcan Wang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongfei Qiao
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yali Li
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
- Jilin Provincial Key Laboratory of Traditional Chinese Medicinal Materials Cultivation and Propagation, Changchun, China
- *Correspondence: Yali Li,
| |
Collapse
|
24
|
Loren P, Lugones Y, Saavedra N, Saavedra K, Páez I, Rodriguez N, Moriel P, Salazar LA. MicroRNAs Involved in Intrinsic Apoptotic Pathway during Cisplatin-Induced Nephrotoxicity: Potential Use of Natural Products against DDP-Induced Apoptosis. Biomolecules 2022; 12:biom12091206. [PMID: 36139046 PMCID: PMC9496062 DOI: 10.3390/biom12091206] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022] Open
Abstract
Cisplatin (cis-diamminedichloroplatinum (II), DDP) is an antineoplastic agent widely used in the treatment of solid tumors because of its extensive cytotoxic activity. However, the main limiting side effect of DDP use is nephrotoxicity, a rapid deterioration in kidney function due to toxic chemicals. Several studies have shown that epigenetic processes are involved in DDP-induced nephrotoxicity. Noncoding RNAs (ncRNAs), a class of epigenetic processes, are molecules that regulate gene expression under physiological and pathological conditions. MicroRNAs (miRNAs) are the most characterized class of ncRNAs and are engaged in many cellular processes. In this review, we describe how different miRNAs regulate some pathways leading to cell death by apoptosis, specifically the intrinsic apoptosis pathway. Accordingly, many classes of natural products have been tested for their ability to prevent DDP-induced apoptosis. The study of epigenetic regulation for underlying cell death is still being studied, which will allow new strategies for the diagnosis and therapy of this unwanted disease, which is presented as a side effect of antineoplastic treatment.
Collapse
Affiliation(s)
- Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
| | - Yuliannis Lugones
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
- Doctoral Programme in Sciences with major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
| | - Isis Páez
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
- Doctoral Programme in Sciences with major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
| | - Nelia Rodriguez
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
- Doctoral Programme in Sciences with major in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
| | - Patricia Moriel
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083970, SP, Brazil
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile
- Correspondence: ; Tel.: +56-452-596-724
| |
Collapse
|
25
|
Domingo IK, Latif A, Bhavsar AP. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. Int J Mol Sci 2022; 23:7227. [PMID: 35806229 PMCID: PMC9266867 DOI: 10.3390/ijms23137227] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cisplatin is a platinum-based chemotherapeutic that has long since been effective against a variety of solid-cancers, substantially improving the five-year survival rates for cancer patients. Its use has also historically been limited by its adverse drug reactions, or cisplatin-induced toxicities (CITs). Of these reactions, cisplatin-induced nephrotoxicity (CIN), cisplatin-induced peripheral neuropathy (CIPN), and cisplatin-induced ototoxicity (CIO) are the three most common of several CITs recognised thus far. While the anti-cancer activity of cisplatin is well understood, the mechanisms driving its toxicities have only begun to be defined. Most of the literature pertains to damage caused by oxidative stress that occurs downstream of cisplatin treatment, but recent evidence suggests that the instigator of CIT development is inflammation. Cisplatin has been shown to induce pro-inflammatory signalling in CIN, CIPN, and CIO, all of which are associated with persisting markers of inflammation, particularly from the innate immune system. This review covered the hallmarks of inflammation common and distinct between different CITs, the role of innate immune components in development of CITs, as well as current treatments targeting pro-inflammatory signalling pathways to conserve the use of cisplatin in chemotherapy and improve long-term health outcomes of cancer patients.
Collapse
Affiliation(s)
| | | | - Amit P. Bhavsar
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (I.K.D.); (A.L.)
| |
Collapse
|
26
|
Wang W, Gu W, He C, Zhang T, Shen Y, Pu Y. Bioactive components of Banxia Xiexin Decoction for the treatment of gastrointestinal diseases based on flavor-oriented analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115085. [PMID: 35150814 DOI: 10.1016/j.jep.2022.115085] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD) was first recorded in a Chinese medical classic, Treatise on Febrile Diseases and Miscellaneous Diseases, which was written in the Eastern Han dynasty of China. This ancient prescription consists of seven kinds of Chinese herbal medicine, namely, Pinellia ternata, Rhizoma Coptidis, Radix scutellariae, Rhizoma Zingiberis, Ginseng, Jujube, and Radix Glycyrrhizaepreparata. In clinic practice, its original application in China mainly has focused on the treatment of chronic gastritis for several hundred years. BXD is also effective in treating other gastrointestinal diseases (GIDs) in modern medical application. Despite available literature support and clinical experience, the treatment mechanisms or their relationships with the bioactive compounds in BXD responsible for its pharmacological actions, still need further explorations in more diversified channels. According to the analysis based on the five-flavor theory of TCM, BXD is traditionally viewed as the most representative prescription for pungent-dispersion, bitter-purgation and sweet-tonification. Consequently, based on the flavor-oriented analysis, the compositive herbs in BXD can be divided into three flavor groups, namely, the pungent, bitter, and sweet groups, each of which has specific active ingredients that are possibly relevant to GID treatment. AIM OF THE REVIEW This paper summarized recent literatures on BXD and its bioactive components used in GID treatment, and provided the pharmacological or chemical basis for the further exploration of the ancient prescription and the relative components. METHOD ology: Relevant literature was collected from various electronic databases such as Pubmed, Web of Science, and China National Knowledge Infrastructure (CNKI). Citations were based on peer-reviewed articles published in English or Chinese during the last decade. RESULTS Multiple components were found in the pungent, bitter, and sweet groups in BXD. The corresponding bioactive components include gingerol, shogaol, stigmasterol, and β-sitosterol in the pungent group; berberine, palmatine, coptisine, baicalein, and baicalin in the bitter group; and ginsenosides, polysaccharides, liquiritin, and glycyrrhetinic acid in the sweet group. These components have been found directly or indirectly responsible for the remarkable effects of BXD on GID. CONCLUSION This review provided some valuable reference to further clarify BXD treatment for GID and their possible material basis, based on the perspective of the flavor-oriented analysis.
Collapse
Affiliation(s)
- Weiwei Wang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiliang Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao He
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yao Shen
- Shanghai Center of Biomedicine Development, Shanghai, 201203, China.
| | - Yiqiong Pu
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
27
|
Zhou X, Zhang K, Liu L, Zhao Q, Huang M, Shao R, Wang Y, Qu B, Wang Y. Anti-fatigue effect from Ginseng Radix et Rhizoma: a suggestive and promising treatment for long COVID. ACUPUNCTURE AND HERBAL MEDICINE 2022; 2:69-77. [PMID: 37808250 PMCID: PMC9407182 DOI: 10.1097/hm9.0000000000000033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/05/2022] [Indexed: 11/26/2022]
Abstract
Two years after the coronavirus disease 2019 (COVID-19) outbreak, an increasing number of patients continue to suffer from long COVID (LC), persistent symptoms, and/or delayed or long-term complications beyond the initial 4 weeks from the onset of symptoms. Constant fatigue is one of the most common LC symptoms, leading to severely reduced quality of life among patients. Ginseng Radix et Rhizoma-known as the King of Herbs in traditional Chinese medicine-has shown clinical anti-fatigue effects. In this review, we summarize the underlying anti-fatigue mechanisms of Ginseng Radix et Rhizoma extracts and their bioactive compounds, with a special focus on anti-viral, immune remodeling, endocrine system regulation, and metabolism, suggesting that Ginseng Radix et Rhizoma is a potentially promising treatment for LC, especially in regard to targeting fatigue.
Collapse
Affiliation(s)
- Xiangda Zhou
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Keying Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lanbo Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qianru Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ming Huang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanyan Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
28
|
Liu C, Zhou S, Bai W, Shi L, Li X. Protective effect of food derived nutrients on cisplatin nephrotoxicity and its mechanism. Food Funct 2022; 13:4839-4860. [PMID: 35416186 DOI: 10.1039/d1fo04391a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platinum-based metal complexes, especially cisplatin (cis-diamminedichloroplatinum II, CDDP), possess strong anticancer properties and a broad anticancer spectrum. However, the clinical application of CDDP has been limited by its side effects including nephrotoxicity, ototoxicity, and neurotoxicity. Furthermore, the therapeutic effects of current clinical protocols are imperfect. Accordingly, it is essential to identify key targets and effective clinical protocols to restrict CDDP-induced nephrotoxicity. Herein, we first analyzed the relevant molecular mechanisms during the process of CDDP-induced nephrotoxicity including oxidative stress, apoptosis, and inflammation. Evidence from current studies was collected and potential targets and clinical protocols are summarized. The evidence indicates an efficacious role of nutrition-based substances in CDDP-induced renal injury.
Collapse
Affiliation(s)
- Chaofan Liu
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Sajin Zhou
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Weibin Bai
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
29
|
Wang Y, Zhang JJ, Hou JG, Li X, Liu W, Zhang JT, Zheng SW, Su FY, Li W. Protective Effect of Ginsenosides from Stems and Leaves of Panax ginseng against Scopolamine-Induced Memory Damage via Multiple Molecular Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1113-1131. [PMID: 35475974 DOI: 10.1142/s0192415x22500458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although growing evidence has shown that ginsenosides from stems and leaves of Panax ginseng (GSLS) exercise a protective impact on the central nervous system, in the model of memory damage induced by scopolamine, it is still rarely reported. Thus, the mechanism of action needs to be further explored. This study was to investigate the effect of GSLS on scopolamine (SCOP)-induced memory damage and the underlying mechanism. Male ICR mice were treated with SCOP (3 mg/kg) for 7 days, with or without GSLS (75 and 150 mg/kg) treatment for 14 days. After GSLS treatment, the memory damage induced by SCOP was significantly ameliorated as shown by the improvement of cholinergic function (AChE and ChAT), brain tissue hippocampus morphology (H&E staining), and oxidative stress (MDA, GSH, and NO). Meanwhile, immunohistochemical assay suggested that GSLS increased the expression of brain-derived neurotrophic factor (BDNF) and Tyrosine Kinase receptor B (TrkB). Further mechanism research indicated that GSLS inhibited the Tau hyperphosphorylation and cell apoptosis by regulating the PI3K/AKT pathway and inhibited neuroinflammation by regulating the NF-κB pathway, thereby exerting a cognitive impairment improvement effect. This work suggested that GSLS could protect against SCOP-induced memory defects possibly through inhibiting oxidative stress, inhibiting neuroinflammation and cell apoptosis.
Collapse
Affiliation(s)
- Ying Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- Intelligent Synthetic Biology Center, Daejeon 34141, Republic of Korea
| | - Xin Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Wei Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Si-Wen Zheng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Feng-Yan Su
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| |
Collapse
|
30
|
Xu H, Liu M, Chen G, Wu Y, Xie L, Han X, Zhang G, Tan Z, Ding W, Fan H, Chen H, Liu B, Zhou Y. Anti-Inflammatory Effects of Ginsenoside Rb3 in LPS-Induced Macrophages Through Direct Inhibition of TLR4 Signaling Pathway. Front Pharmacol 2022; 13:714554. [PMID: 35401188 PMCID: PMC8987581 DOI: 10.3389/fphar.2022.714554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Panax ginseng has therapeutic effects on various inflammation-related diseases. Ginsenoside Rb3 (GRb3), a natural compound with anti-inflammatory and immunomodulatory properties, is one of the main active panaxadiol extracted from Panax ginseng. We explored whether GRb3 inhibited LPS-mediated inflammation through TLR4/NF-κB/MAPK signaling in macrophages. GRb3 attenuated NO and PGE2 production by attenuating iNOS and COX2 expression. GRb3 also suppressed pro-inflammatory cytokines levels, including IL-1β, IL-6, and TNF-α. Moreover, GRb3 administration significantly suppressed NF-κB (p65) nuclear translocation and the phosphorylation levels of p65, IκBα, JNK, p38, and ERK dose-dependently. Molecular docking demonstrated that GRb3 could dock onto the hydrophobic binding site of TLR4/MD2 complex, with a binding energy of −8.79 kcal/mol. Molecular dynamics (MD) displayed stable TLR4-MD2-GRb3 modeling. GRb3 dose-dependently inhibited LPS binding to cell membranes and blocked TLR4 expression. Surface plasmon resonance imaging (SPRi) revealed that GRb3 had an excellent binding affinity to TLR4/MD2 complex. Notably, resatorvid (TAK242), a selective TLR4 inhibitor, did not increase the repressive influence of GRb3 in RAW264.7 macrophages. Moreover, TLR4 overexpression partially reversed the repressive roles of GRb3 on the NF-κB/MAPK pathway and inflammatory mediators. Collectively, our study strongly indicated that GRb3 attenuated LPS-mediated inflammation through direct inhibition of TLR4 signaling. A novel insight into the underlying mechanism of anti-inflammatory effects of GRb3 in macrophages was confirmed.
Collapse
Affiliation(s)
- Honglin Xu
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Min Liu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guanghong Chen
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuting Wu
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Lingpeng Xie
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Han
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhangbin Tan
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wenjun Ding
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Huijie Fan
- TCM Health Construction Department of Yangjiang People’s Hospital, Yangjiang, China
| | - Hongmei Chen
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Yingchun Zhou, ; Bin Liu,
| | - Yingchun Zhou
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Yingchun Zhou, ; Bin Liu,
| |
Collapse
|
31
|
Pavitrakar VN, Mody R, Ravindran S. Protective effects of recombinant human golimumab and pentoxifylline in nephrotoxicity induced by cisplatin. J Biochem Mol Toxicol 2022; 36:e22990. [PMID: 35174923 DOI: 10.1002/jbt.22990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/18/2021] [Accepted: 01/13/2022] [Indexed: 11/11/2022]
Abstract
In cisplatin-induced nephrotoxicity, the significant role of activation of inflammatory pathways has been reported earlier. Studies indicate elevated expression and activity of tumor necrosis factor-α (TNF-α) in both, serum and kidneys of cisplatin-treated animals. Golimumab, an anti-TNF biologic, has been approved for the management of many inflammatory conditions. This experiment was planned and executed to evaluate the impact of golimumab on renal function, inflammation in cisplatin-induced nephrotoxicity in mice, and oxidative stress. Cisplatin (22 mg/kg) as a single intraperitoneal injection was used to induce nephrotoxicity in mice. Golimumab was administered at 24 mg/kg for 7 days by subcutaneous route. Pentoxifylline (PTX) was administered for 7 days (150 mg/kg) as a reference standard. Renal functions, oxidative stress, and inflammation were evaluated on the seventh day. Cisplatin administration in mice caused a significant rise in serum cystatin C, creatinine, blood urea nitrogen, and neutrophil gelatinase-associated lipocalin. A significant rise of urinary clusterin, kidney injury molecule-1, and β-N-acetylglucosaminidase levels was also seen in cisplatin-treated animals. Furthermore, cisplatin-induced nephrotoxicity was associated with a significant increase in oxidative stress and inflammation in serum and kidneys. Golimumab treatment significantly prevented the cisplatin-induced alteration in markers of renal function and renal damage. There was a significant reduction in oxidative stress and inflammation in golimumab-treated animals. Furthermore, the histopathological evaluation also revealed inverted changes in the proximal tubules after golimumab treatment in kidneys after cisplatin toxicity. The standard drug, PTX, also prevented nephrotoxicity caused by cisplatin as indicated by the recovery in renal function, reduction in oxidative stress and inflammation. These results indicate that golimumab was effective in nephrotoxicity induced by cisplatin through inhibition of oxidative stress, and inflammatory response.
Collapse
Affiliation(s)
- Vishal N Pavitrakar
- Biotechnology Division, Lupin Limited, Pune, India.,Faculty of Health Sciences, Symbiosis School of Biological Sciences, Symbiosis International (Deemed) University, Pune, India
| | - Rustom Mody
- Biotechnology Division, Lupin Limited, Pune, India
| | - Selvan Ravindran
- Faculty of Health Sciences, Symbiosis School of Biological Sciences, Symbiosis International (Deemed) University, Pune, India
| |
Collapse
|
32
|
Natural products: potential treatments for cisplatin-induced nephrotoxicity. Acta Pharmacol Sin 2021; 42:1951-1969. [PMID: 33750909 PMCID: PMC8633358 DOI: 10.1038/s41401-021-00620-9] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Cisplatin is a clinically advanced and highly effective anticancer drug used in the treatment of a wide variety of malignancies, such as head and neck, lung, testis, ovary, breast cancer, etc. However, it has only a limited use in clinical practice due to its severe adverse effects, particularly nephrotoxicity; 20%–35% of patients develop acute kidney injury (AKI) after cisplatin administration. The nephrotoxic effect of cisplatin is cumulative and dose dependent and often necessitates dose reduction or withdrawal. Recurrent episodes of AKI result in impaired renal tubular function and acute renal failure, chronic kidney disease, uremia, and hypertensive nephropathy. The pathophysiology of cisplatin-induced AKI involves proximal tubular injury, apoptosis, oxidative stress, inflammation, and vascular injury in the kidneys. At present, there are no effective drugs or methods for cisplatin-induced kidney injury. Recent in vitro and in vivo studies show that numerous natural products (flavonoids, saponins, alkaloids, polysaccharide, phenylpropanoids, etc.) have specific antioxidant, anti-inflammatory, and anti-apoptotic properties that regulate the pathways associated with cisplatin-induced kidney damage. In this review we describe the molecular mechanisms of cisplatin-induced nephrotoxicity and summarize recent findings in the field of natural products that undermine these mechanisms to protect against cisplatin-induced kidney damage and provide potential strategies for AKI treatment.
Collapse
|
33
|
Wan Y, Wang J, Xu JF, Tang F, Chen L, Tan YZ, Rao CL, Ao H, Peng C. Panax ginseng and its ginsenosides: potential candidates for the prevention and treatment of chemotherapy-induced side effects. J Ginseng Res 2021; 45:617-630. [PMID: 34764717 PMCID: PMC8569258 DOI: 10.1016/j.jgr.2021.03.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy-induced side effects affect the quality of life and efficacy of treatment of cancer patients. Current approaches for treating the side effects of chemotherapy are poorly effective and may cause numerous harmful side effects. Therefore, developing new and effective drugs derived from natural non-toxic compounds for the treatment of chemotherapy-induced side effects is necessary. Experiments in vivo and in vitro indicate that Panax ginseng (PG) and its ginsenosides are undoubtedly non-toxic and effective options for the treatment of chemotherapy-induced side effects, such as nephrotoxicity, hepatotoxicity, cardiotoxicity, immunotoxicity, and hematopoietic inhibition. The mechanism focus on anti-oxidation, anti-inflammation, and anti-apoptosis, as well as the modulation of signaling pathways, such as nuclear factor erythroid-2 related factor 2 (Nrf2)/heme oxygenase-1 (HO-1), P62/keap1/Nrf2, c-jun N-terminal kinase (JNK)/P53/caspase 3, mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinases (ERK), AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase kinase 4 (MKK4)/JNK, and phosphatidylinositol 3-kinase (PI3K)/AKT. Since a systemic review of the effect and mechanism of PG and its ginsenosides on chemotherapy-induced side effects has not yet been published, we provide a comprehensive summarization with this aim and shed light on the future research of PG.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADM, Adriamycin
- ALT, alanine aminotransferase
- AMO, Atractylodes macrocephala volatile oil
- AMPK, AMP-activated protein kinase
- ARE, antioxidant response element
- AST, aspartate aminotransferase
- BMNC, bone marrow nucleated cells
- CIA, chemotherapy-induced hair loss
- CK, compound K
- CP, cisplatin
- CY, cyclophosphamide
- CYP2E1, Cytochrome P450 E1
- Chemotherapy
- DAC, doses of docetaxel, doxorubicin as well as cyclophosphamide
- ERG, enzyme-treated eRG
- ERK, extracellular signal-regulated kinases
- FBG, fermented black ginseng
- FRG, probiotic-fermented eRG
- FRGE, fermented red ginseng extract
- GM-CSF, granulocyte macrophage colony-stimulating factor
- Ginsenosides
- HEI-OC1, House Ear Institute-Organ of Corti 1
- HO-1, heme oxygenase-1
- HSPCS, haematopoietic stem and progenitor cells
- IL, interleukin
- JNK, c-jun N-terminal kinase
- KG-KH, the mixture of ginsenosides Rk3 and Rh4
- LLC-PK1, porcine renal proximal epithelial tubular
- LSK, Lin−Sca-1+c-kit+
- MAPK, mitogen-activated protein kinase
- MDA, malonaldehyde
- MEK, mitogen activated protein kinase
- MKK4, mitogen activated protein kinase kinase 4
- Mechanism
- NF-κB, nuclear factor-kappa B p65
- NQO, NAD (P) H quinone oxidoreductase
- Nrf2, nuclear factor erythroid related factor 2
- PG
- PG, Panax ginseng
- PGFR, PG flower
- PGLF, PG leaf
- PGRT, PG root
- PGS, PG total saponins
- PGSD, PG seeds
- PGSM, PG stem
- PI3K, phosphatidylinositol 3-kinase
- PPD, protopanaxadiol
- PPT, protopanaxatriol
- Pharmacological effects
- RG, red ginseng
- RGE, red ginseng extract
- ROS, reactive oxygen species
- SREBP-1, sterol regulatory element binding protein 1
- Side effects
- TNF-α, tumor necrosis factor-α
- eRG, 50% ethanol-extracted RG
- mTOR, mammalian target of rapamycin
- wRG, water-extracted RG
Collapse
Affiliation(s)
- Yan Wan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-feng Xu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-zhu Tan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao-long Rao
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
34
|
Leta B, Kenenisa C, Wondimnew T, Sime T. Evaluation of Renoprotective Effects of Our Locally Grown Green Coffee Beans against Cisplatin-Induced Nephrotoxicity in Swiss Albino Mice. Int J Nephrol 2021; 2021:2805068. [PMID: 34676116 PMCID: PMC8526242 DOI: 10.1155/2021/2805068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/18/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Nephrotoxicity is the most common and severe side effect of cisplatin. Cisplatin causes nephrotoxicity through free radical production and debilitating cellular antioxidant capacity. Coffee is a commonly consumed drink and its ingredients have antioxidant roles that could bring benefits to patients affected by nephrotoxicity. Thus, the present study aimed to investigate the renoprotective effects of our locally grown green coffee beans against cisplatin-induced nephrotoxicity in Swiss albino mice. METHODS The posttest only control group design was employed on a total of thirty male Swiss albino mice. The mice were divided into five groups: group I (normal control group) received distilled water; group II (negative control group) received distilled water; and groups III-V (treatment groups) received 100, 200, and 300 mg/kg BW/day of green coffee bean extract for 14 days, respectively. Nephrotoxicity was induced in groups II-V by a single intraperitoneal injection of cisplatin (7.5 mg/kg). All mice were sacrificed after 14 days and blood was drawn to evaluate kidney function tests (serum creatinine and serum blood urea nitrogen). Besides, body weight, relative kidney weight, and kidney histopathology were investigated. RESULT Our results showed that treatment of cisplatin alone (group II mice) significantly increased serum creatinine, serum blood urea nitrogen, relative kidney weight, and pathological damage to the kidney with a decrease in final body weight. However, low-dose green coffee beans (group III), medium-dose green coffee beans (group IV), and high-dose green coffee beans (group V) mice showed a significant dose-dependent decrease in serum creatinine, serum blood urea nitrogen, and relative kidney weight. Furthermore, the dose-dependent treatment with green coffee bean extract prevented the decrease in body weight gain and pathological damage to the kidney in mice. CONCLUSION Our locally grown green coffee beans brought a dose-dependent ameliorative effect and a promising preventive approach against cisplatin-induced kidney damage in mice.
Collapse
Affiliation(s)
- Bati Leta
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Chala Kenenisa
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Tesaka Wondimnew
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Tariku Sime
- Department of Biomedical Sciences, Faculty of Medical Sciences, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| |
Collapse
|
35
|
Zhang JJ, Zhou YD, Liu YB, Wang JQ, Li KK, Gong XJ, Lin XH, Wang YP, Wang Z, Li W. Protective Effect of 20(R)-Ginsenoside Rg3 Against Cisplatin-Induced Renal Toxicity via PI3K/AKT and NF-[Formula: see text]B Signaling Pathways Based on the Premise of Ensuring Anticancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1739-1756. [PMID: 34461812 DOI: 10.1142/s0192415x21500828] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although the protective effect of ginsenoside on cisplatin-induced renal injury has been extensively studied, whether ginsenoside interferes with the antitumor effect of cisplatin has not been confirmed. In this paper, we verified the main molecular mechanism of 20(R)-ginsenoside Rg3 (R-Rg3) antagonizing cisplatin-induced acute kidney injury (AKI) through the combination of in vivo and in vitro models. It is worth mentioning that the two cell models of HK-2 and HepG2 were used simultaneously for the first time to explore the effect of the activation site of tumor-associated protein p53 on apoptosis and tumor suppression. The results showed that a single injection of cisplatin (20 mg/kg) led to weight loss, the kidney index of the mice increased, and creatinine (CRE) and blood urea nitrogen (BUN) levels in mice sharply increased. Continuous administration of R-Rg3 at doses of 10 and 20 mg/kg for 10 days could significantly alleviate this symptom. Similarly, R-Rg3 treatment reduced oxidative stress damage caused by cisplatin. Moreover, R-Rg3 could observably reduce the apoptosis and inflammatory infiltration of renal tubular cells induced by cisplatin. We used western blotting analysis to demonstrate that R-Rg3 restored cisplatin-induced AKI might be related to PI3K/AKT and NF-[Formula: see text]B mediated apoptosis and inflammation pathways. In the meantime, we also verified that R-Rg3 could activate different sites of p53 to control renal cell apoptosis induced by cisplatin without affecting its antitumor effect.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yan-Dan Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yong-Bo Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jian-Qiang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Ke-Ke Li
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiao-Jie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiang-Hui Lin
- Liaoning XIFENG Pharmaceutical Group Co., Ltd., Huanren 117200, P. R. China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China.,Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| |
Collapse
|
36
|
Liu MY, Liu F, Gao YL, Yin JN, Yan WQ, Liu JG, Li HJ. Pharmacological activities of ginsenoside Rg5 (Review). Exp Ther Med 2021; 22:840. [PMID: 34149886 PMCID: PMC8210315 DOI: 10.3892/etm.2021.10272] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Ginseng, a perennial plant belonging to genus Panax, has been widely used in traditional herbal medicine in East Asia and North America. Ginsenosides are the most important pharmacological component of ginseng. Variabilities in attached positions, inner and outer residues and types of sugar moieties may be associated with the specific pharmacological activities of each ginsenoside. Ginsenoside Rg5 (Rg5) is a minor ginsenoside synthesized during ginseng steaming treatment that exhibits superior pharmaceutical activity compared with major ginsenosides. With high safety and various biological functions, Rg5 may act as a potential therapeutic candidate for diverse diseases. To date, there have been no systematic studies on the activity of Rg5. Therefore, in this review, all available literature was reviewed and discussed to facilitate further research on Rg5.
Collapse
Affiliation(s)
- Ming-Yang Liu
- Department of Immunity, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fei Liu
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan-Li Gao
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia-Ning Yin
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei-Qun Yan
- Department of Tissue Engineering, School of Pharmaceutical Sciences in Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian-Guo Liu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai-Jun Li
- Department of Immunity, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
37
|
Sha JY, Li JH, Zhou YD, Yang JY, Liu W, Jiang S, Wang YP, Zhang R, Di P, Li W. The p53/p21/p16 and PI3K/Akt signaling pathways are involved in the ameliorative effects of maltol on D-galactose-induced liver and kidney aging and injury. Phytother Res 2021; 35:4411-4424. [PMID: 34028092 DOI: 10.1002/ptr.7142] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 03/17/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022]
Abstract
Successive evidence has established that maltol, a flavor-enhancing agent, could provide resistance to oxidative stress-induced tissue injury in various animal models though its benefits for aging-induced liver and kidney injuries are still undetermined. In the present work, for demonstrating maltol's ameliorative effect and probable mechanism against aging-induced liver and kidney injuries, D-galactose (D-Gal)-induced animal in vivo and HEK293 cells in vitro models were established and results demonstrated that long-term D-Gal treatment increases the accumulation of advanced glycation end products (AGEs) in liver and kidney tissues, mitigates cell viability, and arrests the cycle. Interestingly, 4-weeks maltol treatment at 50 and 100 mg/kg activated aging-associated proteins including p53, p21, and p16 followed by inhibiting malondialdehyde (MDA)'s over-production and increasing the levels of antioxidant enzymes. Therefore, decreases in cytochrome P450 E1 (CYP2E1) and 4-hydroxydecene (4-HNE)'s immunofluorescence expression levels are confirmed. Furthermore, maltol improved oxidative stress injury by activating the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. In conclusion, the purpose of the present study was to estimate the mechanistic insights into maltol's role as an antioxidant in liver and kidney cell senescence and injury, which will reflect potential of therapeutic strategy for antiaging and aging-related disease treatment.
Collapse
Affiliation(s)
- Ji-Yue Sha
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jian-Hao Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,Plant Chemistry Laboratory, Chinese Institute of Jilin Ginseng, Changchun, China
| | - Yan-Dan Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jia-Yu Yang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Wei Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Rui Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Peng Di
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| |
Collapse
|
38
|
Gao C, Liu C, Chen Y, Wang Q, Hao Z. Protective effects of natural products against drug-induced nephrotoxicity: A review in recent years. Food Chem Toxicol 2021; 153:112255. [PMID: 33989732 DOI: 10.1016/j.fct.2021.112255] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
Drug-induced nephrotoxicity (DIN) is a major cause of kidney damage and is associated with high mortality and morbidity, which limits the clinical use of certain therapeutic or diagnostic agents, such as antineoplastic drugs, antibiotics, immunosuppressive agents, non-steroidal anti-inflammatory drugs (NSAIDs), and contrast agents. However, in recent years, a number of studies have shown that many natural products (NPs), including phytochemicals, various plants extracts, herbal formulas, and NPs derived from animals, confer protective effects against DIN through multi-targeting therapeutic mechanisms, such as inhibition of oxidative stress, inflammation, apoptosis, fibrosis, and necroptosis, regulation of autophagy, maintenance of cell polarity, etc., by regulating multiple signaling pathways and novel molecular targets. In this review, we summarize and discuss the protective effects and mechanisms underlying the action of NPs against DIN found in recent years, which will contribute to the development of promising renal protective agents.
Collapse
Affiliation(s)
- Chen Gao
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Chang Liu
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yuwei Chen
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Qingtao Wang
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Zhihui Hao
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
39
|
Abstract
Cisplatin has been a mainstay of cancer chemotherapy since the 1970s. Despite its broad anticancer potential, its clinical use has regularly been constrained by kidney toxicities. This review details those biochemical pathways and metabolic conversions that underlie the kidney toxicities. A wide range of redox events contribute to the eventual physiological consequences of drug activities.
Collapse
|
40
|
Wei XM, Jiang S, Li SS, Sun YS, Wang SH, Liu WC, Wang Z, Wang YP, Zhang R, Li W. Endoplasmic Reticulum Stress-Activated PERK-eIF2α-ATF4 Signaling Pathway is Involved in the Ameliorative Effects of Ginseng Polysaccharides against Cisplatin-Induced Nephrotoxicity in Mice. ACS OMEGA 2021; 6:8958-8966. [PMID: 33842766 PMCID: PMC8027996 DOI: 10.1021/acsomega.0c06339] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 05/25/2023]
Abstract
Although previous studies have reported that saponins (ginsenosides, the major active and most representative ingredients in Panax ginseng C.A. Meyer) exerted a good ameliorative effect on cisplatin (CP)-induced acute kidney injury in animal models, little attention has been paid to a large number of polysaccharides isolated and purified from ginseng. This work aimed to investigate the protective effect and the possible molecular mechanism of ginseng polysaccharide (WGP) on CP-induced kidney toxicology in mice. The results from biomarker analysis including serum creatinine (CRE) and blood urea nitrogen (BUN) confirmed the protective effect of WGP at 200 and 400 mg/kg on CP-induced renal-toxicology. We found that WGP reduces the apoptosis of kidney cells by inhibiting endoplasmic reticulum (ER) stress caused by CP, which is manifested by increased phosphorylation of PERK. In addition, the apoptosis-associated with caspase 3 activation in renal cells induced by CP was inhibited after administration of WGP, and the phosphorylation levels of PI3K and AKT were also reduced significantly. We also demonstrated that after exposure to CP, the unfolded protein response signaling pathway PERK-eIF2α-ATF4 axis was significantly activated, manifested by increased phosphorylation of eIF2α and increased expression of ATF4 and CHOP. Interestingly, the WGP administration improves this situation. Furthermore, the supplement of WGP inhibited the overexpression of nuclear factor-kappa B p65 (NF-κB p65) and tumor necrosis factor-α (TNF-α) caused by CP exposure. In short, for the first time, our findings indicated that WGP could effectively prevent CP-induced ER stress, inflammation, and apoptosis in renal cells, in part, by regulating the PI3K/AKT and PERK-eIF2α-ATF4 signaling pathways.
Collapse
Affiliation(s)
- Xiao-meng Wei
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Shuang Jiang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Shan-shan Li
- Institute
of Special Wild Economic Animals and Plant, Chinese Academy of Agricultural Sciences, Changchun 132109, China
| | - Yin-shi Sun
- Institute
of Special Wild Economic Animals and Plant, Chinese Academy of Agricultural Sciences, Changchun 132109, China
| | - Shi-han Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Wen-cong Liu
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Zi Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Ying-ping Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Rui Zhang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Wei Li
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| |
Collapse
|
41
|
Yoo S, Park BI, Kim DH, Lee S, Lee SH, Shim WS, Seo YK, Kang K, Lee KT, Yim SV, Soung DY, Kim BH. Ginsenoside Absorption Rate and Extent Enhancement of Black Ginseng (CJ EnerG) over Red Ginseng in Healthy Adults. Pharmaceutics 2021; 13:pharmaceutics13040487. [PMID: 33918329 PMCID: PMC8067055 DOI: 10.3390/pharmaceutics13040487] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 11/17/2022] Open
Abstract
Red ginseng (RG) and black ginseng (BG, CJ EnerG) were prepared from fresh ginseng using one and nine cycles of steaming and drying, respectively. This process reduces the molecular weight (MW) of ginsenoside-active compounds in ginseng by removing sugar moieties from their dammaranes. We compared the pharmacokinetic characteristics of ginsenosides between BG comprising mainly low-MW ginsenosides (Rg3, Rg5, Rk1, and Rh1) and RG that predominantly contains high-MW ginsenosides (Rb1, Rb2, Rc, Rd, Re, and Rg1). The safety profiles and tolerability were also studied using a randomized, double-blind, single-dose, crossover clinical trial. A combination of Rb1, Rg1, and Rg3, well-known representative and functional RG components, exhibited a 1 h faster absorption rate (Tmax) and 58% higher exposure (24 h area under the concentration–time curve, AUC24) in BG than in RG. Furthermore, the combination of Rg3, Rg5, and Rk1, the major and most efficient components in BG, displayed 824% higher absorption (AUC24) in BG than in RG. The total ginsenoside showed a 5 h rapid intestinal absorption (Tmax) and 79% greater systemic exposure (AUC24) in BG than in RG. No clinically significant findings were observed in terms of safety or tolerability. Thus, BG extract was more effective than RG extract.
Collapse
Affiliation(s)
- Saebyul Yoo
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.Y.); (D.-h.K.)
| | - Bom-I Park
- Food Research Institutes, CJ CheilJedang, Suwon 16495, Korea; (B.-I.P.); (Y.K.S.); (K.K.)
| | - Do-hyun Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.Y.); (D.-h.K.)
| | - Sooyoung Lee
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.L.); (K.-T.L.)
| | - Seung-hoon Lee
- Department of Statistics, lnha University, Incheon 22212, Korea;
| | - Wang-Seob Shim
- Kyung Hee Drug Analysis Center, College of Pharmacy, Medical Center, Kyung Hee University, Seoul 02447, Korea;
| | - Yong Ki Seo
- Food Research Institutes, CJ CheilJedang, Suwon 16495, Korea; (B.-I.P.); (Y.K.S.); (K.K.)
| | - Kimoon Kang
- Food Research Institutes, CJ CheilJedang, Suwon 16495, Korea; (B.-I.P.); (Y.K.S.); (K.K.)
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.L.); (K.-T.L.)
- Kyung Hee Drug Analysis Center, College of Pharmacy, Medical Center, Kyung Hee University, Seoul 02447, Korea;
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Sung-Vin Yim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
| | - Do Yu Soung
- Food Research Institutes, CJ CheilJedang, Suwon 16495, Korea; (B.-I.P.); (Y.K.S.); (K.K.)
- Correspondence: (D.Y.S.); (B.-H.K.)
| | - Bo-Hyung Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (S.Y.); (D.-h.K.)
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (D.Y.S.); (B.-H.K.)
| |
Collapse
|
42
|
Yousof Ali M, Jannat S, Mizanur Rahman M. Ginsenoside derivatives inhibit advanced glycation end-product formation and glucose-fructose mediated protein glycation in vitro via a specific structure-activity relationship. Bioorg Chem 2021; 111:104844. [PMID: 33798848 DOI: 10.1016/j.bioorg.2021.104844] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/29/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022]
Abstract
Ginseng (Panax ginseng and red ginseng) extract has been reported to inhibit the formation of advanced glycation end-products (AGEs); however, the potential inhibitory activity of its major constituents (ginsenosides) against AGE formation is still unknown. In the present study, we investigated the inhibitory effect of ginsenoside derivatives on AGE formation. Herein, we assessed the activity of 22 ginsenosides, most of which significantly inhibited fluorescent AGE formation. Notably, ginsenoside Rh2, ginsenoside Rh1, and compound K exhibited the most potent AGE inhibitory potential with IC50 values of 3.38, 8.42, and 10.85 µM, respectively. The structure- activity relationship revealed that the presence of sugar moieties, hydroxyl groups, and their linkages, and the stereostructure of the ginsenoside skeleton played an important role in the inhibition of AGE formation. Furthermore, the inhibitory activity of the most active ginsenoside Rh2 on fructose-glucose-mediated protein glycation and oxidation of bovine serum albumin (BSA) was explored. Rh2 (0.1-12.5 µM) inhibited the formation of fluorescent AGE and non-fluorescent AGE, as well as the level of fructosamine and prevented protein oxidation by decreasing protein carbonyl formation and protein thiol group modification. Rh2 also suppressed the formation of the β-cross amyloid structure of BSA. Ginsenosides might be promising new anti-glycation agents for the prevention of diabetic complications via inhibition of AGE formation and oxidation-dependent protein damage.
Collapse
Affiliation(s)
- Md Yousof Ali
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemistry and Biochemistry, Faculty of Arts and Science, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec, Canada; Department of Biology, Faculty of Arts and Science, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec, Canada; Centre for Structural and Functional Genomic, Dept. of Biology, Faculty of Arts and Science, Concordia University, 7141 Sherbrooke St. W., Montreal, QC, Canada.
| | - Susoma Jannat
- Department of Biochemistry and Molecular Biology, University of Calgary, T2N 1N4 Alberta, Canada
| | - M Mizanur Rahman
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
43
|
Kim HJ, Kim SJ, Woo CW, Kim ST, Im M, Park SK, Kim JY, Yoo HJ, Woo DC, Kim JK. Treatment of chemotherapy-induced cachexia with BST204: a multimodal validation study. Metabolomics 2021; 17:36. [PMID: 33738589 DOI: 10.1007/s11306-021-01781-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/26/2021] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Chemotherapy is a major etiology of cachexia. Ginseng products are known to have various anti-cachectic and health-promoting effects, such as inhibiting inflammation and promoting energy production. In particular, BST204, purified ginseng dry extract, contains multiple ginsenosides that can reduce chemotherapy-related fatigue and toxicity. OBJECTIVES To investigate the effects of BST204 on the alleviation of chemotherapy-induced cachexia using a multimodal approach. METHODS In a CT26 mouse syngeneic colon cancer model, cachexia was predominantly induced by chemotherapy with 5-fluorouracil (5-FU) than by tumor growth. BST204 at a dose of 100 or 200 mg/kg was administered to 5-FU-treated mice. RESULTS BST204 significantly mitigated the decrease in tumor-excluded body weight (change in 5-FU group and BST204 groups: - 13% vs. - 6% on day 7; - 30% vs. - 20% on day 11), muscle volume (- 19% vs. - 11%), and fat volume (- 91% vs. - 56%). The anti-cachectic effect of BST204 was histologically demonstrated by an improved balance between muscle regeneration and degeneration and a decrease in muscle cross-sectional area reduction. CONCLUSION Chemotherapy-induced cachexia was biochemically and metabolically characterized by activated inflammation, enhanced oxidative stress, increased protein degradation, decreased protein stabilization, reduced glucose-mediated energy production, and deactivated glucose-mediated biosynthesis. These adverse effects were significantly improved by BST204 treatment. Overall, our multimodal study demonstrated that BST204 could effectively alleviate chemotherapy-induced cachexia.
Collapse
Affiliation(s)
- Ho-Jin Kim
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Su Jung Kim
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chul-Woong Woo
- MR Core Laboratory, Asan Medical Center, Asan Medical Institute of Convergence and Technology,, Seoul, Republic of Korea
| | - Sang-Tae Kim
- MR Core Laboratory, Asan Medical Center, Asan Medical Institute of Convergence and Technology,, Seoul, Republic of Korea
| | - Minju Im
- GREEN CROSS Wellbeing Co., Ltd, Seongnam, Republic of Korea
| | - Sun Kyu Park
- GREEN CROSS Wellbeing Co., Ltd, Seongnam, Republic of Korea
| | - Jeom-Yong Kim
- GREEN CROSS Wellbeing Co., Ltd, Seongnam, Republic of Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Dong-Cheol Woo
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- MR Core Laboratory, Asan Medical Center, Asan Medical Institute of Convergence and Technology,, Seoul, Republic of Korea.
| | - Jeong Kon Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
He S, Lyu F, Lou L, Liu L, Li S, Jakowitsch J, Ma Y. Anti-tumor activities of Panax quinquefolius saponins and potential biomarkers in prostate cancer. J Ginseng Res 2021; 45:273-286. [PMID: 33841008 PMCID: PMC8020356 DOI: 10.1016/j.jgr.2019.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 10/28/2019] [Accepted: 12/30/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Prostate carcinoma is the second most common cancer among men worldwide. Developing new therapeutic approaches and diagnostic biomarkers for prostate cancer (PC) is a significant need. The Chinese herbal medicine Panax quinquefolius saponins (PQS) have been reported to show anti-tumor effects. We hypothesized that PQS exhibits anti-cancer activity in human PC cells and we aimed to search for novel biomarkers allowing early diagnosis of PC. METHODS We used the human PC cell line DU145 and the prostate epithelial cell line PNT2 to perform cell viability assays, flow cytometric analysis of the cell cycle, and FACS-based apoptosis assays. Microarray-based gene expression analysis was used to display specific gene expression patterns and to search for novel biomarkers. Western blot and quantitative real-time PCR were performed to demonstrate the expression levels of multiple cancer-related genes. RESULTS Our data showed that PQS inhibited the viability of DU145 cells and induced cell cycle arrest at the G1 phase. A significant decrease in DU145 cell invasion and migration were observed after 24 h treatment by PQS. PQS up-regulated the expression levels of p21, p53, TMEM79, ACOXL, ETV5, and SPINT1 while it down-regulated the expression levels of bcl2, STAT3, FANCD2, DRD2, and TMPRSS2. CONCLUSION PQS promoted cells apoptosis and inhibited the proliferation of DU145 cells, which suggests that PQS may be effective for treating PC. TMEM79 and ACOXL were expressed significantly higher in PNT2 than in DU145 cells and could be novel biomarker candidates for PC diagnosis.
Collapse
Key Words
- ACOXL, Acyl-CoA oxidase-like protein
- Chinese medicinal herbs
- DRD2, dopamine receptor D2
- ETV5, ETS variant 5
- FACS, fluorescence-activated cell sorting
- FANCD2, fanconi anemia group D2
- PC, prostate cancer
- PQS, Panax quinquefolius saponins
- Panax quinquefolius
- Potential biomarkers
- Prostate cancer cells
- SPINT1, serine peptidase inhibitor Kunitz type 1
- STAT3, signal transducer and activator of transcription 3
- TCM, Traditional Chinese Medicine
- TMEM79, transmembrane protein 79
- TMPRSS2, transmembrane protease serine 2
- bcl2, B-cell lymphoma 2
- p21, cyclin-dependent kinase inhibitor p21
- p53, tumor suppressor p53
- qRT-PCR, quantitative real-time PCR
- saponins
Collapse
Affiliation(s)
- Shan He
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Fangqiao Lyu
- Department of Cell Biology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Lixia Lou
- The Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Songlin Li
- Department of Pharmaceutical Analysis and Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing, China
| | - Johannes Jakowitsch
- Department of Internal Medicine, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Yan Ma
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Moreno KGT, Gasparotto Junior A, Dos Santos AC, Palozi RAC, Guarnier LP, Marques AAM, Romão PVM, Lorençone BR, Cassemiro NS, Silva DB, Tirloni CAS, de Barros ME. Nephroprotective and antilithiatic activities of Costus spicatus (Jacq.) Sw.: Ethnopharmacological investigation of a species from the Dourados region, Mato Grosso do Sul State, Brazil. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113409. [PMID: 32979411 DOI: 10.1016/j.jep.2020.113409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Costus spicatus (Jacq.) Sw., also known as "cana-do-brejo," is a species that is widely used in Brazilian traditional medicine for the treatment of kidney diseases. However, no studies have evaluated its nephroprotective and antilithiatic effects. AIM To investigate nephroprotective and antilithiatic effects of C. spicatus in a preclinical model of acute kidney injury (AKI) and in vitro nephrolithiasis. MATERIALS AND METHODS C. spicatus leaves were collected directly from the natural environment in the Dourados region, Mato Grosso do Sul State, Brazil. The ethanol-soluble fraction of C. spicatus (ESCS) was obtained by infusion. Phytochemical characterization was performed by liquid chromatography coupled to diode array detector and mass spectrometer (LC-DAD-MS). We assessed whether ESCS has acute or prolonged diuretic activity. The nephroprotective effects of ESCS were evaluated in a model of AKI that was induced by glycerol (10 ml/kg, intramuscularly) in Wistar rats. Different doses of ESCS (30, 100, and 300 mg/kg) were administered orally for 5 days before the induction of AKI. Urinary parameters were measured on days 1, 3, 5, and 7. Twenty-four hours after the last urine collection, blood samples were obtained for the biochemical analysis. Blood pressure levels, renal vascular reactivity, renal tissue redox status, and histopathological changes were measured. Antilithiatic effects were evaluated by in vitro crystallization. Calcium oxalate precipitation was induced by sodium oxalate in urine samples with ESCS at 0.05, 0.5, and 5 mg/ml. RESULTS From LC-DAD-MS analyses, flavonoids, saponins and other phenolic compounds were determined in the composition of ESCS. Significant reductions of the excretion of urinary total protein, creatinine, sodium, and potassium were observed in the AKI group, with significant histopathological damage (swelling, vacuolization, necrosis, and inflammatory infiltration) in the proximal convoluted tubule. Treatment with ESCS exerted a significant nephroprotective effect by increasing the urinary excretion of total protein, urea, creatinine, sodium, potassium, calcium, and chloride. All of the groups that were treated with ESCS exhibited a reduction of histopathological lesions and significant modulation of the tissue redox state. We also observed a concentration-dependent effect of ESCS on the crystallization of urinary crystals, with reductions of the size and proportion of monohydrated crystals. CONCLUSION The data suggest that C. spicatus has nephroprotective and antilithiatic effects, suggesting possible effectiveness in its traditional use.
Collapse
Affiliation(s)
- Karyne Garcia Tafarelo Moreno
- Laboratório de Urinálise, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Arquimedes Gasparotto Junior
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil.
| | - Ariany Carvalho Dos Santos
- Laboratório de Histopatologia, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Rhanany Alan Calloi Palozi
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Lucas Pires Guarnier
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Aline Aparecida Macedo Marques
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Paulo Vitor Moreira Romão
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Bethânia Rosa Lorençone
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Nadla Soares Cassemiro
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal Do Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Denise Brentan Silva
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal Do Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Cleide Adriane Signor Tirloni
- Laboratório de Farmacologia Cardiovascular- LaFaC, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Márcio Eduardo de Barros
- Laboratório de Urinálise, Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| |
Collapse
|
46
|
Protective Effects of Shenfuyixin Granule on H 2O 2-Induced Apoptosis in Neonatal Rat Cardiomyocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6654457. [PMID: 33564318 PMCID: PMC7867454 DOI: 10.1155/2021/6654457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 12/01/2022]
Abstract
Shenfuyixin granule (SFYXG, i.e., Xinshuaikang granule) is a prescription, commonly used in the clinical experience, which plays a significant role in the treatment of heart failure. The purpose of this present research was to investigate the protective effect of SFYXG, and the mechanism about anti-H2O2-induced oxidative stress and apoptosis in the neonatal rat cardiomyocytes. Myocardial cells, as is well known, were divided into 4 groups: normal, model, SFYXG, and coenzyme Q10 group, respectively. Cells viability was determined by MTT assay. Flow cytometry and AO/EB staining were implemented to test the apoptosis rate and intracellular reactive oxygen species (ROS) level. Mitochondrion membrane potential (MMP) was evaluated by JC-1 fluorescence probe method. The myocardial ultrastructure of mitochondrion was measured by electron microscope. The related mRNA expression levels of Bax, Bcl-2, Caspase-3, caspase-8, and caspase-9 were detected by real-time polymerase chain reaction (PCR). Also, the expression levels of Bax and Bcl-2 protein were detected by Western blot, and the expression levels of caspase-3, caspase-8, and caspase-9 protein were tested by caspase-Glo®3 Assay, caspase-Glo®8 Assay, and caspase-Glo®9 Assay, respectively. GAPDH was used as the internal reference gene/protein. The results revealed that SFYXG (0.5 mg/ml) raised the viability of myocardial cell, weakened the apoptosis rate and ROS level, corrected the mitochondrion membrane potential stability, and improved cell morphology and ultrastructure of myocardial mitochondrion. Furthermore, SFYXG upregulated the antiapoptosis gene of Bcl-2, but downregulated the proapoptosis genes of Bax, caspase-3, and caspase-9. In conclusion, SFYXG could appear to attenuate myocardial injury by its antioxidative and antiapoptosis effect.
Collapse
|
47
|
Liu YQ, Wang XL, He DH, Cheng YX. Protection against chemotherapy- and radiotherapy-induced side effects: A review based on the mechanisms and therapeutic opportunities of phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153402. [PMID: 33203590 DOI: 10.1016/j.phymed.2020.153402] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/29/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Although great achievements have been made in the field of cancer therapy, chemotherapy and radiotherapy remain the mainstay cancer therapeutic modalities. However, they are associated with various side effects, including cardiocytotoxicity, nephrotoxicity, myelosuppression, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, mucositis, and alopecia, which severely affect the quality of life of cancer patients. Plants harbor a great chemical diversity and flexible biological properties that are well-compatible with their use as adjuvant therapy in reducing the side effects of cancer therapy. PURPOSE This review aimed to comprehensively summarize the molecular mechanisms by which phytochemicals ameliorate the side effects of cancer therapies and their potential clinical applications. METHODS We obtained information from PubMed, Science Direct, Web of Science, and Google scholar, and introduced the molecular mechanisms by which chemotherapeutic drugs and irradiation induce toxic side effects. Accordingly, we summarized the underlying mechanisms of representative phytochemicals in reducing these side effects. RESULTS Representative phytochemicals exhibit a great potential in reducing the side effects of chemotherapy and radiotherapy due to their broad range of biological activities, including antioxidation, antimutagenesis, anti-inflammation, myeloprotection, and immunomodulation. However, since a majority of the phytochemicals have only been subjected to preclinical studies, clinical trials are imperative to comprehensively evaluate their therapeutic values. CONCLUSION This review highlights that phytochemicals have interesting properties in relieving the side effects of chemotherapy and radiotherapy. Future studies are required to explore the clinical benefits of these phytochemicals for exploitation in chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Liu
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Xiao-Lu Wang
- Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, China
| | - Dan-Hua He
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yong-Xian Cheng
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
48
|
Ryu J, Yoon J, Lee YW. Kinetic study of the thermal conversion of ginsenosides using lumped groups in steaming, hydrothermal reactions, and CO2-assisted hydrothermal reactions. J Supercrit Fluids 2021. [DOI: 10.1016/j.supflu.2020.105041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
49
|
Ameliorative Effect of Linalool in Cisplatin-Induced Nephrotoxicity: The Role of HMGB1/TLR4/NF-κB and Nrf2/HO1 Pathways. Biomolecules 2020; 10:biom10111488. [PMID: 33126443 PMCID: PMC7693927 DOI: 10.3390/biom10111488] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The monoterpene linalool is a well-known essential oil component produced by several aromatic plants. Cisplatin is a widely used anticancer drug that produces many side effects, particularly nephrotoxicity. Here, we aimed to inspect linalool's protective activity against cisplatin-induced nephrotoxicity and explore part of the underlying mechanisms. METHODS Male Wistar rats were given linalool (50 and 100 mg/kg/day orally) for 15 days; then challenged with cisplatin (8 mg/kg) on the 12th day. Renal function parameters, oxidative stress, inflammatory and apoptotic markers, and toll-like receptor pathway gene, and protein expressions were investigated. Histopathology, immunohistochemistry, and cell-line mediated cytotoxicity assays were conducted. RESULTS Linalool ameliorated kidney function after cisplatin challenge and managed all oxidation system parameters including GSH, SOD, CAT, MDA, NADPH, and particularly the Nrf2-mediated pathway markers. Linalool decreased TLR4, MYD88 and TRIF gene and protein expressions; diminished related inflammatory mediators such as TNF-α, IL-1β, IL-6, and NF-κB; and down-regulated HMBG1. Linalool mitigated cisplatin-induced apoptotic markers such as caspase 3, caspase 9, and Bax expression, and boosted the anti-apoptotic Bcl2 expression. Linalool potentiated the cytotoxic effect of cisplatin when investigated on HeLa and PC3 human cancer cell lines. CONCLUSION Linalool could protect against cisplatin-induced kidney function and tissue damage.
Collapse
|
50
|
Liu W, Leng J, Hou JG, Jiang S, Wang Z, Liu Z, Gong XJ, Chen C, Wang YP, Li W. Saponins derived from the stems and leaves of Panax ginseng attenuate scrotal heat-induced spermatogenic damage via inhibiting the MAPK mediated oxidative stress and apoptosis in mice. Phytother Res 2020; 35:311-323. [PMID: 32767418 DOI: 10.1002/ptr.6801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Heat stress (HS) reaction is a stress response caused by adverse conditions. Currently, the incidence of reproductive malignancies particularly in males has been constantly increasing. This work investigated the effects of saponins derived from the stems and leaves of Panax ginseng (GSLS) on testicular injury induced by scrotal hyperthermia in mice. GSLS (150, 300 mg/kg) were administered intragastrically to mice for 14 days, then exposed to a single scrotal heat treatment at 43°C for 18 min on seventh day. HS induced a significant loss of multinucleate giant cells, desquamation of germ cells in destructive seminiferous tubules. Moreover, HS reduced the serum testosterone, testicular tissue superoxide dismutase activity and glutathione (GSH) content, while significantly enhanced the production of malondialdehyde (p < .05). GSLS exhibited the protective potential against HS-induced injury not only by modulating Bcl-2 family and caspase protease family, but also by suppressing the protein levels of heme oxygenase-1 (HO-1), heat shock protein 70 (HSP70), hypoxia inducible factor-1α (HIF-1α) and activation of Mitogen-activated protein kinase (MAPK) signaling pathways (p < .05). In conclusion, we clearly demonstrated that GSLS exhibited a significant protective effect against HS-induced testicular dysfunction, mainly the inhibition of oxidative stress associated apoptosis partly via regulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Wei Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jing Leng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jin-Gang Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,Intelligent Synthetic Biology Center, Daejeon, Republic of Korea
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Xiao-Jie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, China
| |
Collapse
|