1
|
Tasheva K, Sulikovska I, Georgieva A, Djeliova V, Lozanova V, Vasileva A, Ivanov I, Denev P, Lazarova M, Vassileva V, Petkova-Kirova P. Phytochemical Profile, Antioxidant Capacity and Anticancer Potential of Water Extracts from In Vitro Cultivated Salvia aethiopis. Molecules 2025; 30:1427. [PMID: 40286005 PMCID: PMC11990555 DOI: 10.3390/molecules30071427] [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: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025] Open
Abstract
Salvia aethiopis L. (Mediterranean sage) is a medicinal plant known for its rich phenolic content and different therapeutic properties. This study evaluated the phytochemical composition, antioxidant capacity and anticancer potential of water extracts from in vitro cultivated S. aethiopis. The extract exhibited a high total polyphenol (110.03 ± 0.7 mg GAE/g) and flavonoid (7.88 ± 0.25 mg QE/g) content, along with a strong oxygen radical absorbance capacity (an ORAC value of 3677.9 ± 24.8 µmol TE/g). LC-HRMS analysis identified 21 bioactive compounds, including salvianic acid C, rosmarinic acid, salvianolic acid K and various organic acids. A cytotoxicity evaluation using the Neutral Red Uptake assay showed that the extract had a low toxicity to non-cancerous BALB/3T3 cells. An antiproliferative activity assessment via the MTT assay revealed selective cytotoxicity against Hep G2 hepatocellular carcinoma cells (IC50 = 353.8 ± 21.8 µg/mL) and lung (A549) and prostate (PC-3) carcinoma cell lines. Migration assays and cytopathological evaluations confirmed the significant inhibition of cancer cell proliferation, the suppression of migration and G2/M cell cycle arrest. Flow cytometry revealed considerable increases in apoptotic and necrotic cell populations following treatment with S. aethiopis extract. These findings showed the potential of S. aethiopis as a promising source of bioactive compounds with antioxidant and anticancer properties, supporting its further exploration for therapeutic applications.
Collapse
Affiliation(s)
- Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Inna Sulikovska
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (I.S.); (A.G.)
| | - Ani Georgieva
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (I.S.); (A.G.)
| | - Vera Djeliova
- Department of Molecular Biology of Cell Cycle, Institute of Molecular Biology “Acad. R. Tsanev”, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Vesela Lozanova
- Department of Medical Chemistry and Biochemistry, Medical University–Sofia, 1431 Sofia, Bulgaria; (V.L.); (A.V.); (I.I.)
| | - Anelia Vasileva
- Department of Medical Chemistry and Biochemistry, Medical University–Sofia, 1431 Sofia, Bulgaria; (V.L.); (A.V.); (I.I.)
| | - Ivaylo Ivanov
- Department of Medical Chemistry and Biochemistry, Medical University–Sofia, 1431 Sofia, Bulgaria; (V.L.); (A.V.); (I.I.)
| | - Petko Denev
- Laboratory of Biologically Active Substances, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 4000 Plovdiv, Bulgaria;
| | - Maria Lazarova
- Department of Synaptic Signaling and Communication, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (M.L.); (P.P.-K.)
| | - Valya Vassileva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Polina Petkova-Kirova
- Department of Synaptic Signaling and Communication, Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (M.L.); (P.P.-K.)
| |
Collapse
|
2
|
Li HB, Huang L, Ni JY, Lin RY, Xi SY. Traditional Chinese medicine in the treatment of adverse reactions after TACE for primary hepatic carcinoma: Effect, mechanism, and potential advantages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156244. [PMID: 39556987 DOI: 10.1016/j.phymed.2024.156244] [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: 03/21/2024] [Revised: 09/19/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Primary hepatic carcinoma is one of the most common malignant tumors. China is a major country for liver cancer, accounting for about 50 % of the patients worldwide. Although there are a variety of treatments for primary hepatic carcinoma, chemotherapy remains an important method, and transcatheter arterial chemoembolization (TACE) is a commonly used local chemotherapy. Currently, there are no effective therapeutic measures to target adverse reactions generated after chemoembolization. A new approach is needed to alleviate post-TACE syndrome. Clinical and experimental studies have shown that traditional Chinese medicine can reduce adverse reactions and improve clinical efficacy when combined with primary hepatic carcinoma treatment. This suggests that traditional Chinese medicine plays an important and irreplaceable role in alleviating adverse reactions after TACE. However, there is still a need for high-quality experimental and clinical studies to obtain evidence of effective treatment.
Collapse
Affiliation(s)
- Huai-Bang Li
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Lin Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jia-Yu Ni
- Department of Clinical Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Ruo-Yao Lin
- Department of Clinical Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Sheng-Yan Xi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen 361102, China; Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen 361101, China; School of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang 43900, Malaysia.
| |
Collapse
|
3
|
Arto C, Rusu EC, Clavero-Mestres H, Barrientos-Riosalido A, Bertran L, Mahmoudian R, Aguilar C, Riesco D, Chicote JU, Parada D, Martínez S, Sabench F, Richart C, Auguet T. Metabolic profiling of tryptophan pathways: Implications for obesity and metabolic dysfunction-associated steatotic liver disease. Eur J Clin Invest 2024; 54:e14279. [PMID: 38940215 DOI: 10.1111/eci.14279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/12/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND AND AIMS The rise in obesity highlights the need for improved therapeutic strategies, particularly in addressing metabolic dysfunction-associated steatotic liver disease (MASLD). We aim to assess the role of tryptophan metabolic pathways in the pathogenesis of obesity and in the different histological stages of MASLD. MATERIALS AND METHODS We used ultra-high performance liquid chromatography to quantify circulating levels of 15 tryptophan-related metabolites from the kynurenine, indole and serotonin pathways. A cohort of 76 subjects was analysed, comprising 18 subjects with normal weight and 58 with morbid obesity, these last being subclassified into normal liver (NL), simple steatosis (SS) and metabolic dysfunction-associated steatohepatitis (MASH). Then, we conducted gene expression analysis of hepatic IDO-1 and kynyrenine-3-monooxygenase (KMO). RESULTS Key findings in obesity revealed a distinct metabolic signature characterized by a higher concentration of different kynurenine-related metabolites, a decrease in indole-3-acetic acid and indole-3-propionic acid, and an alteration in the serotonin pathway. Elevated tryptophan levels were associated with MASLD presence (37.659 (32.577-39.823) μM of tryptophan in NL subjects; 41.522 (38.803-45.276) μM in patients with MASLD). Overall, pathway fluxes demonstrated an induction of tryptophan catabolism via the serotonin pathway in SS subjects and into the kynurenine pathway in MASH. We found decreased IDO-1 and KMO hepatic expression in NL compared to SS. CONCLUSIONS We identified a distinctive metabolic signature in obesity marked by changes in tryptophan catabolic pathways, discernible through altered metabolite profiles. We observed stage-specific alterations in tryptophan catabolism fluxes in MASLD, highlighting the potential utility of targeting these pathways in therapeutic interventions.
Collapse
Affiliation(s)
- Carmen Arto
- Servei Medicina Interna, Hospital Sant Pau i Santa Tecla de Tarragona, Tarragona, Spain
| | - Elena Cristina Rusu
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Helena Clavero-Mestres
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Andrea Barrientos-Riosalido
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Laia Bertran
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Razieh Mahmoudian
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Carmen Aguilar
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - David Riesco
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Javier Ugarte Chicote
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Servei Anatomia Patològica, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - David Parada
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Servei Anatomia Patològica, Hospital Sant Joan de Reus, Avinguda Doctor Josep Laporte, Reus, Spain
| | - Salomé Martínez
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Servei Anatomia Patològica, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Fàtima Sabench
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Departament de Medicina i Cirurgia, Servei de Cirurgia, Hospital Sant Joan de Reus, URV, IISPV, Avinguda Doctor Josep Laporte, Reus, Spain
| | - Cristóbal Richart
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Teresa Auguet
- Departament de Medicina i Cirurgia, Grup de Recerca GEMMAIR (AGAUR)-Medicina Aplicada (URV), Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| |
Collapse
|
4
|
Niu C, Zhang J, Okolo PI. The possible pathogenesis of liver fibrosis: therapeutic potential of natural polyphenols. Pharmacol Rep 2024; 76:944-961. [PMID: 39162986 DOI: 10.1007/s43440-024-00638-w] [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: 03/13/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
Liver fibrosis is the formation of a fibrous scar resulting from chronic liver injury, independently from etiology. Although many of the mechanical details remain unknown, activation of hepatic stellate cells (HSCs) is a central driver of liver fibrosis. Extracellular mechanisms such as apoptotic bodies, paracrine stimuli, inflammation, and oxidative stress are critical in activating HSCs. The potential for liver fibrosis to reverse after removing the causative agent has heightened interest in developing antifibrotic therapies. Polyphenols, the secondary plant metabolites, have gained attention because of their health-beneficial properties, including well-recognized antioxidant and anti-inflammatory activities, in the setting of liver fibrosis. In this review, we present an overview of the mechanisms underlying liver fibrosis with a specific focus on the activation of resident HSCs. We highlight the therapeutic potential and promising role of natural polyphenols to mitigate liver fibrosis pathogenesis, focusing on HSCs activation. We also discuss the translational gap from preclinical findings to clinical treatments involved in natural polyphenols in liver fibrosis.
Collapse
Affiliation(s)
- Chengu Niu
- Internal medicine residency program, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY, 14621, USA.
| | - Jing Zhang
- Rainier Springs Behavioral Health Hospital, 2805 NE 129th St, Vancouver, WA, 98686, USA
| | - Patrick I Okolo
- Division of Gastroenterology, Rochester General Hospital, Rochester, NY, 14621, USA
| |
Collapse
|
5
|
Mor A, Tankiewicz-Kwedlo A, Ciwun M, Lewkowicz J, Pawlak D. Kynurenines as a Novel Target for the Treatment of Inflammatory Disorders. Cells 2024; 13:1259. [PMID: 39120289 PMCID: PMC11311768 DOI: 10.3390/cells13151259] [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: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
This review discusses the potential of targeting the kynurenine pathway (KP) in the treatment of inflammatory diseases. The KP, responsible for the catabolism of the amino acid tryptophan (TRP), produces metabolites that regulate various physiological processes, including inflammation, cell cycle, and neurotransmission. These metabolites, although necessary to maintain immune balance, may accumulate excessively during inflammation, leading to systemic disorders. Key KP enzymes such as indoleamine 2,3-dioxygenase 1 (IDO1), indoleamine 2,3-dioxygenase 2 (IDO2), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO) have been considered promising therapeutic targets. It was highlighted that both inhibition and activation of these enzymes may be beneficial, depending on the specific inflammatory disorder. Several inflammatory conditions, including autoimmune diseases, for which modulation of KP activity holds therapeutic promise, have been described in detail. Preclinical studies suggest that this modulation may be an effective treatment strategy for diseases for which treatment options are currently limited. Taken together, this review highlights the importance of further research on the clinical application of KP enzyme modulation in the development of new therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Adrian Mor
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Anna Tankiewicz-Kwedlo
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Marianna Ciwun
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Janina Lewkowicz
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland;
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| |
Collapse
|
6
|
Cui H, Tang Y, Yang C, Deng H, Chen L, Fan X, Zhu L, Liu Y, Zhao Z, Su T. Meroterpenoids from the marine-derived fungus Aspergillus terreus GZU-31-1 exerts anti-liver fibrosis effects by targeting the Nrf2 signaling in vitro. PHYTOCHEMISTRY 2024; 219:113983. [PMID: 38215814 DOI: 10.1016/j.phytochem.2024.113983] [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: 08/18/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Six undescribed meroterpenoids aspertermeroterpenes A-F and four known analogues were isolated from the marine-derived fungus Aspergillus terreus GZU-31-1. Their structures were elucidated based on spectroscopic methods and electronic circular dichroism calculations. All meroterpenoids possessed the unique acetyl group at C-11, and also aspertermeroterpene A featured the rare C-14 decarboxylated in DMOA meroterpenoids. In the bioassays, aspermeroterpene B exhibited a potent inhibitory effect on the activation of hepatic stellate cells at the concentration of 5 μM via targeting the Nrf2 signaling. This is the first time reported that aspermeroterpene B as a previously undescribed carbon skeleton of meroterpenoid possessed anti-liver fibrosis effect.
Collapse
Affiliation(s)
- Hui Cui
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yuqian Tang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; South China Sea Institute of Oceanology, University of Chinese Academy of Sciences, Guangzhou, 510301, China
| | - Chunfang Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huimei Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lei Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xueying Fan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Liping Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yena Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Zhongxiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Tao Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Wang Y, Ji X, Zhao M, Li J, Yin H, Jin J, Zhao L. Modulation of tryptophan metabolism via AHR-IL22 pathway mediates the alleviation of DSS-induced colitis by chitooligosaccharides with different degrees of polymerization. Carbohydr Polym 2023; 319:121180. [PMID: 37567716 DOI: 10.1016/j.carbpol.2023.121180] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 08/13/2023]
Abstract
Oral administration of chitooligosaccharides (COS) has been reported to alleviate colitis in mice. However, the mechanism of action of COS with specific polymerization degree on gut inflammation and metabolism remains unclear. This study aimed to investigate the effects of chitobiose (COS2), chitotetraose (COS4), and chitohexaose (COS6) on colitis, and to elucidate their underlying mechanisms. COS2, COS4, and COS6 were able to significantly alleviate colonic injury and inflammation levels. COS6 has the best anti-inflammatory effect. Furthermore, COS6 could down-regulate the level of indoleamine-2,3-dioxygenase1 (IDO1) and restore the levels of indole, indoleacetic-3-acid (IAA), and indole-3-carbaldehyde (I3A) in the cecum of chronic colitis mice (p < 0.05), thereby regulating tryptophan metabolism. In the aromatic hydrocarbon receptor-IL-22 (AHR-IL-22) pathway, although there were differences between chronic colitis and acute colitis mice, COS intervention could restore the AHR-IL-22 pathway to normal, promote the expression of MUC2, and repair the intestinal mucosal barrier. In conclusion, the results of this study suggested that COS had a good inhibitory effect on IDO1 under inflammation and the changes of AHR and IL-22 levels at different stages of disease development. This provides new insights into the potential use of COS as a functional food for improving intestinal inflammation and metabolism.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoguo Ji
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China
| | - Mengyao Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China
| | - Juan Li
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Jiayang Jin
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai 200237, China.
| | - Liming Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China.
| |
Collapse
|
8
|
Wang Y, Song J, Yu K, Nie D, Zhao C, Jiao L, Wang Z, Zhou L, Wang F, Yu Q, Zhang S, Wen Z, Wu J, Wang CY, Wang DW, Cheng J, Zhao C. Indoleamine 2,3-Dioxygenase 1 Deletion-Mediated Kynurenine Insufficiency Inhibits Pathological Cardiac Hypertrophy. Hypertension 2023; 80:2099-2111. [PMID: 37485661 DOI: 10.1161/hypertensionaha.122.20809] [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: 12/15/2022] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Aberrant amino acid metabolism is implicated in cardiac hypertrophy, while the involvement of tryptophan metabolism in pathological cardiac hypertrophy remains elusive. Herein, we aimed to investigate the effect and potential mechanism of IDO1 (indoleamine 2,3-dioxygenase) and its metabolite kynurenine (Kyn) on pathological cardiac hypertrophy. METHODS Transverse aortic constriction was performed to induce cardiac hypertrophy in IDO1-knockout (KO) mice and AAV9-cTNT-shIDO1 mice. Liquid chromatography-mass spectrometry was used to detect the metabolites of tryptophan-Kyn pathway. Chromatin immunoprecipitation assay and dual luciferase assay were used to validate the binding of protein and DNA. RESULTS IDO1 expression was upregulated in both human and murine hypertrophic myocardium, alongside with increased IDO1 activity and Kyn content in transverse aortic constriction-induced mice's hearts using liquid chromatography-mass spectrometry analysis. Myocardial remodeling and heart function were significantly improved in transverse aortic constriction-induced IDO1-KO mice, but were greatly exacerbated with subcutaneous Kyn administration. IDO1 inhibition or Kyn addition confirmed the alleviation or aggravation of hypertrophy in cardiomyocyte treated with isoprenaline, respectively. Mechanistically, IDO1 and metabolite Kyn contributed to pathological hypertrophy via the AhR (aryl hydrocarbon receptor)-GATA4 (GATA binding protein 4) axis. CONCLUSIONS This study demonstrated that IDO1 deficiency and consequent Kyn insufficiency can protect against pathological cardiac hypertrophy by decreasing GATA4 expression in an AhR-dependent manner.
Collapse
Affiliation(s)
- Yinhui Wang
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Jia Song
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.S.)
| | - Kun Yu
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Daan Nie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (D.N.)
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China (D.N.)
| | - Chengcheng Zhao
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Liping Jiao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China (L.J.)
| | - Ziyi Wang
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Ling Zhou
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Feng Wang
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Qilin Yu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Q.Y., S.Z., C.-Y.W.)
| | - Shu Zhang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Q.Y., S.Z., C.-Y.W.)
| | - Zheng Wen
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Junfang Wu
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Cong-Yi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Q.Y., S.Z., C.-Y.W.)
| | - Dao Wen Wang
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Jia Cheng
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| | - Chunxia Zhao
- Department of Internal Medicine, Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Y.W., K.Y., Chengcheng Zhao, Z. Wang, L.Z., F.W., Z. Wen, J.W., D.W.W., J.C., Chunxia Zhao)
| |
Collapse
|
9
|
Yang J, Wei W, Gao C, Song W, Gao C, Chen X, Liu J, Guo L, Liu L, Wu J. Efficient production of salvianic acid A from L-dihydroxyphenylalanine through a tri-enzyme cascade. BIORESOUR BIOPROCESS 2023; 10:31. [PMID: 38647923 PMCID: PMC10992476 DOI: 10.1186/s40643-023-00649-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/31/2023] [Indexed: 04/25/2024] Open
Abstract
Salvianic acid A (SAA), used for treating cardiovascular and cerebrovascular diseases, possesses several pharmacological properties. However, the current methods for the enzymatic synthesis of SAA show low efficiency. Here, we constructed a three-enzyme cascade pathway in Escherichia coli BL21 (DE3) to produce SAA from L-dihydroxyphenylalanine (L-DOPA). The phenylpyruvate reductase (LaPPR) from Lactobacillus sp. CGMCC 9967 is a rate-limiting enzyme in this process. Therefore, we employed a mechanism-guided protein engineering strategy to shorten the transfer distances of protons and hydrides, generating an optimal LaPPR mutant, LaPPRMu2 (H89M/H143D/P256C), with a 2.8-fold increase in specific activity and 9.3-time increase in kcat/Km value compared to that of the wild type. Introduction of the mutant LaPPRMu2 into the cascade pathway and the optimization of enzyme levels and transformation conditions allowed the obtainment of the highest SAA titer (82.6 g L-1) ever reported in vivo, good conversion rate (91.3%), excellent ee value (99%) and the highest productivity (6.9 g L-1 h-1) from 90 g L-1 L-DOPA in 12 h. This successful strategy provides a potential new method for the industrial production of SAA.
Collapse
Affiliation(s)
- Jiahui Yang
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Wanqing Wei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Changzheng Gao
- Department of Cardiology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Wei Song
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Cong Gao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiulai Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jia Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Liang Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Liming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jing Wu
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
10
|
Lv L, Wang D, Yin J, Yang T, Huang B, Cao Y, Lu J. Downregulation of miR-20b-5p Contributes to the Progression of Liver Fibrosis via the STAT3 Signaling Pathway In Vivo and In Vitro. Dig Dis Sci 2023; 68:487-496. [PMID: 35947307 DOI: 10.1007/s10620-022-07660-z] [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] [Received: 03/04/2022] [Accepted: 08/02/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Activated hepatic stellate cells (HSCs) are primarily involved in liver fibrosis and portal hypertension (PHT). We aimed to investigate the effect of miR-20b-5p on HSCs, liver fibrosis, and PHT. METHODS MiR-20b-5p expression in HSCs and in mouse liver fibrosis was determined by qPCR. Further, the effects of miR-20b-5p mimic on HSCs migration, proliferation, and apoptosis were investigated in vitro. A dual-luciferase reporter assay was performed to confirm the direct interaction between miR-20b-5p and STAT3. In vivo, mouse liver fibrosis was established by common bile duct ligation and intervened by agomiR-20b-5p. Sirius red staining and hydroxyproline content were used to evaluate collagen deposition. The α-SMA expression in the liver was detected by IHC and Western blotting. The STAT3 signaling pathway and its downregulated cytokines as well as portal pressure and angiogenesis were explored. RESULTS MiR-20b-5p was significantly downregulated during HSCs activation and in mouse liver fibrosis. The functional analyses demonstrated that miR-20b-5p inhibited cell proliferation, activation, and promoted apoptosis in HSCs in vitro. Moreover, miR-20b-5p regulated STAT3 expression by binding to the 3'UTR of its miRNA directly. Overexpression of miR-20b-5p facilitated HSC activation and proliferation by inhibiting the STAT3 signaling pathway. MiR-20b-5p overexpression suppressed the STAT3 and its downstream cytokines and ameliorated liver fibrosis in mice. The intra- and inter-hepatic angiogenesis were also effectively inhibited. The inhibition of liver fibrosis and neoangiogenesis contributed to the decrease of portal pressure. CONCLUSIONS MiR-20b-5p plays an important role in the fibrosis and angiogenesis of liver fibrosis by targeting the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ling Lv
- Department of Disease Control and Prevention, Tangdu Hospital, Fourth Military Medical University, 569 Xin Si Road, Xi'an, 710038, China
| | - Dong Wang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| | - Jikai Yin
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Tao Yang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Huang
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanlong Cao
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianguo Lu
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Wang C, Su Z, Xu J, Ko C. Danshensu attenuated lipopolysaccharide-induced LX-2 and T6 cells activation through regulation of ferroptosis. Food Sci Nutr 2023; 11:344-349. [PMID: 36655094 PMCID: PMC9834887 DOI: 10.1002/fsn3.3065] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 01/21/2023] Open
Abstract
Liver fibrosis and cirrhosis are primarily caused by the activation of hepatic stellate cells (HSCs), regardless of their etiology. Collagen type I (collagen I) and connective tissue growth factor (CTGF) is produced more readily by activated HSCs. Consequently, identifying the molecular and cellular mechanisms responsible for HSCs activation is essential to better understand its mechanism of action and therapeutic potential. Cell death is caused by iron-dependent lipid peroxidation during ferroptosis. Ferroptosis plays an important role in the survival of activated HSCs and could contribute to the development of innovative prevention and treatment strategies for liver fibrosis. Danshensu (Dan) is a pure molecule extracted from the Salvia miltiorrhiza herb that protects against liver damage. However, Dan's effect on attenuating HSCs activation by regulating ferroptosis remains unclear. The results of this study indicated that lipopolysaccharide (LPS)-induced LX-2 and T6 cells activation occurs through the upregulation of collagen I, CTGF, Gpx4, and SLC7A11. Interestingly, Dan attenuated LPS-induced liver fibrosis in those cells by upregulating collagen I, CTGF, Gpx4, and SLC7A11 and by increasing lipid reactive oxygen species accumulation. Furthermore, the results also showed that the ferroptosis inhibitor liproxstatin attenuated the overproduction of lipid reactive oxygen species in LPS-activated LX-2 cells. We conclude that Dan attenuates LPS-induced HSC activation during liver fibrosis by regulating ferroptosis in LX-2 and T6 cells.
Collapse
Affiliation(s)
- Changting Wang
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Zhiming Su
- Department of General SurgeryThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Jian‐Hua Xu
- Department of Tumor SurgeryThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Chih‐Yuan Ko
- Department of Clinical NutritionThe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- School of Public HealthFujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
12
|
Ameliorative effects of Danshensu from the functional food Salvia miltiorrhiza against arsenic trioxide-induced cardiac toxicity in vivo and in vitro: Involvement of inhibiting the AKT/IKK/NF-κB signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
13
|
Zhao Q, Bai J, Chen Y, Liu X, Zhao S, Ling G, Jia S, Zhai F, Xiang R. An optimized herbal combination for the treatment of liver fibrosis: Hub genes, bioactive ingredients, and molecular mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115567. [PMID: 35870684 DOI: 10.1016/j.jep.2022.115567] [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: 05/22/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is a chronic liver disease that can lead to cirrhosis, liver failure, and hepatocellular carcinoma, and it is associated with long-term adverse outcomes and mortality. As a primary resource for complementary and alternative medicine, traditional Chinese medicine (TCM) has accumulated a large number of effective formulas for the treatment of liver fibrosis in clinical practice. However, studies on how to systematically optimize TCM formulas are still lacking. AIM OF THE REVIEW To provide a methodological reference for the systematic optimization of TCM formulae against liver fibrosis and explored the underlying molecular mechanisms; To provide an efficient method for searching for lead compounds from natural sources and developing from herbal medicines; To enable clinicians and patients to make more reasonable choices and promote the effective treatment toward those patients with liver fibrosis. MATERIALS AND METHODS TCM formulas related to treating liver fibrosis were collected from the Web of Science, PubMed, the China National Knowledge Infrastructure (CNKI), Wan Fang, and the Chinese Scientific Journals Database (VIP). Furthermore, the TCM compatibility patterns were mined using association analysis. The core TCM combinations were found by designing an optimized formulas algorithm. Finally, the hub target proteins, potential molecular mechanisms, and active compounds were explored through integrative pharmacology and docking-based inverse virtual screening (IVS) approaches. RESULTS We found that the herbs for reinforcing deficiency, activating blood, removing blood stasis, and clearing heat were the basis of TCM formulae patterns. Furthermore, the combination of Salviae Miltiorrhizae (Salvia miltiorrhiza Bunge; Chinese salvia/Danshen), Astragali Radix (Astragalus membranaceus (Fisch.) Bunge; Astragalus/Huangqi), and Radix Bupleuri (Bupleurum chinense DC.; Bupleurum/Chaihu) was identified as core groups. A total of six targets (TNF, STAT3, EGFR, IL2, ICAM1, PTGS2) play a pivotal role in TCM-mediated liver fibrosis inhibition. (-)-Cryptotanshinone, Tanshinaldehyde, Ononin, Thymol, Daidzein, and Formononetin were identified as active compounds in TCM. And mechanistically, TCM could affect the development of liver fibrosis by regulating inflammation, immunity, angiogenesis, antioxidants, and involvement in TNF, MicroRNAs, Jak-STAT, NF-kappa B, and C-type lectin receptors (CLRs) signaling pathways. Molecular docking results showed that key components had good potential to bind to the target genes. CONCLUSION In summary, this study provides a methodological reference for the systematic optimization of TCM formulae and exploration of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Bai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yiwei Chen
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xin Liu
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shangfeng Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Guixia Ling
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shubing Jia
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Fei Zhai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Rongwu Xiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China; Liaoning Professional Technology Innovation Center on Medical Big Data and Artificial Intelligence, Shenyang, 110016, China.
| |
Collapse
|
14
|
Zhou P, An B, Zhang X, Lv J, Lin B. Therapeutic effect and mechanism of danshensu on coronary heart disease using liquid chromatography combined with mass spectrometry metabolomics. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1208:123400. [PMID: 35917776 DOI: 10.1016/j.jchromb.2022.123400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/17/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022]
Abstract
Metabolomics can discover the biomarkers and metabolic pathways, provides the possibility for insights into the pharmacological action and mechanism of natural products. The therapeutic effect and mechanism of danshensu (DSS) on total metabolic pathways has not been well investigated. The aim of this study was to explore the disturbed endogenous biomarkers and metabolic pathways reflecting the pharmacological activity of DSS, and mechanism of action of DSS using comprehensive metabolome analysis based on high-throughput metabolomics technology combined with ultra-high performance liquid chromatography (UPLC) coupled with quadrupole tandem time-of-flight mass spectrometry (Q-TOF-MS) and pattern recognition method. Through the changes of the overall metabolic profile and the related biomarkers, the intervention effect of natural product danshensu (DSS) treatment on CHD model rats was revealed. The results showed that after the model replication was established, the metabolic profile was clearly separated, and a total of 26 potential biomarkers were screened out, and involving 8 metabolic pathways. After different doses of DSS solution were given, a total of 20 biomarkers could be significantly regulated, mainly involving primary bile acid biosynthesis, glycerophospholipid metabolism, and lipid metabolism. It showed UPLC-MS-based metabolomics can be used for discovering potential biomarkers and metabolic pathways of CHD, and to further understand and dissecting pharmacological effects and mechanisms of natural products via metabolomics techniques.
Collapse
Affiliation(s)
- Peng Zhou
- School of Continuing Education, Heilongjiang University of Traditional Chinese Medicine, Heping Road No. 24, Xiangfang District, Harbin City, Heilongjiang Province, China.
| | - Baisong An
- Drug Safety Evaluation Center of Heilongjiang University of Traditional Chinese Medicine, Heping Road No. 24, Xiangfang District, Harbin City, Heilongjiang Province, China
| | - Xiaolei Zhang
- School of Continuing Education, Heilongjiang University of Traditional Chinese Medicine, Heping Road No. 24, Xiangfang District, Harbin City, Heilongjiang Province, China
| | - Jiming Lv
- Drug Purchasing Center, Daqing Traditional Chinese Medicine Hospital, Health Road No. 8, Saertu District, Daqing City, Heilongjiang Province, China
| | - Baisong Lin
- Department of Blood Transfusion, Jiamusi Central Hospital, 256 Zhongshan Street, Xiangyang District, Jiamusi City, Heilongjiang Province, China.
| |
Collapse
|
15
|
Zhang XL, Zhang XY, Ge XQ, Liu MX. Mangiferin prevents hepatocyte epithelial-mesenchymal transition in liver fibrosis via targeting HSP27-mediated JAK2/STAT3 and TGF-β1/Smad pathway. Phytother Res 2022; 36:4167-4182. [PMID: 35778992 DOI: 10.1002/ptr.7549] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 12/11/2022]
Abstract
Hepatocytes has been confirmed to undergo EMT and can be converted into myofibroblasts during hepatic fibrogenesis. However, the mechanism of hepatocyte EMT regulation in hepatic fibrosis, particularly through HSP27 (human homologue of rodent HSP25), remains unclear. Mangiferin (MAN), a compound extracted from Mangifera indica L, has been reported to attenuate liver injury. This study aimed to investigate the mechanisms underlying HSP27 inhibition and the anti-fibrotic effect of MAN in liver fibrosis. Our results revealed that the expression of HSP27 was remarkably increased in the liver tissues of patients with liver cirrhosis and CCl4 -induced fibrotic rats. However, HSP27 shRNA treatment significantly alleviated fibrosis. Furthermore, MAN was found to inhibit CCl4 - and TGF-β1-induced liver fibrosis and reduced hepatocyte EMT. More importantly, MAN decreased HSP27 expression to suppress the JAK2/STAT3 pathway, and subsequently blocked TGF-β1/Smad signaling, which were consistent with its protection against CCl4 -induced EMT and liver fibrosis. Together, these results suggest that HSP27 may play a crucial role in hepatocyte EMT and liver fibrosis by activating JAK2/STAT3 signaling and TGF-β1/Smad pathway. The suppression of HSP27 expression by MAN may be a novel strategy for attenuating the hepatocyte EMT in liver fibrosis.
Collapse
Affiliation(s)
- Xiao-Ling Zhang
- College of Pharmacy, Nantong University, Nantong, PR China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiao-Yan Zhang
- Department of Pharmacology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, PR China
| | - Xiao-Qun Ge
- Department of Pharmacology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, PR China
| | - Ming-Xuan Liu
- College of Pharmacy, Nantong University, Nantong, PR China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| |
Collapse
|
16
|
Qin LJ, Lu Z, Zheng ZH, Zhong XH. Research progress of natural products interfering with cell signaling pathway in liver fibrosis. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:15-23. [PMID: 33779429 DOI: 10.1080/10286020.2021.1883003] [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: 09/08/2020] [Accepted: 01/25/2021] [Indexed: 06/12/2023]
Abstract
Liver cirrhosis and hepatocellular carcinoma are the late stage of liver fibrosis. How to early use drugs to intervene in liver fibrosis is a prerequisite for the reversal of liver fibrosis. This paper mainly introduces a cell signaling transduction pathway in liver fibrosis and the intervention of natural products in order to provide theoretical basis for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Li-Jing Qin
- Undergraduate Class of Medical Imaging, Jilin Medical University, Jilin 132013, China
| | - Zhang Lu
- Undergraduate Class of Medical Imaging, Jilin Medical University, Jilin 132013, China
| | - Zhong-Hua Zheng
- Department of Pathology, Jilin Medical University, Jilin 132013, China
| | - Xiu-Hong Zhong
- Department of Pathology, Jilin Medical University, Jilin 132013, China
| |
Collapse
|
17
|
Wang R, Wang Z, Sun R, Fu R, Sun Y, Zhu M, Geng Y, Gao D, Tian X, Zhao Y, Yao J. Activation of TAF9 via Danshensu-Induced Upregulation of HDAC1 Expression Alleviates Non-alcoholic Fatty Liver Disease. Front Pharmacol 2021; 12:775528. [PMID: 34925033 PMCID: PMC8678612 DOI: 10.3389/fphar.2021.775528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/22/2021] [Indexed: 11/17/2022] Open
Abstract
Fatty acid β-oxidation is an essential pathogenic mechanism in nonalcoholic fatty liver disease (NAFLD), and TATA-box binding protein associated factor 9 (TAF9) has been reported to be involved in the regulation of fatty acid β-oxidation. However, the function of TAF9 in NAFLD, as well as the mechanism by which TAF9 is regulated, remains unclear. In this study, we aimed to investigate the signaling mechanism underlying the involvement of TAF9 in NAFLD and the protective effect of the natural phenolic compound Danshensu (DSS) against NAFLD via the HDAC1/TAF9 pathway. An in vivo model of high-fat diet (HFD)-induced NAFLD and a palmitic acid (PA)-treated AML-12 cell model were developed. Pharmacological treatment with DSS significantly increased fatty acid β-oxidation and reduced lipid droplet (LD) accumulation in NAFLD. TAF9 overexpression had the same effects on these processes both in vivo and in vitro. Interestingly, the protective effect of DSS was markedly blocked by TAF9 knockdown. Mechanistically, TAF9 was shown to be deacetylated by HDAC1, which regulates the capacity of TAF9 to mediate fatty acid β-oxidation and LD accumulation during NAFLD. In conclusion, TAF9 is a key regulator in the treatment of NAFLD that acts by increasing fatty acid β-oxidation and reducing LD accumulation, and DSS confers protection against NAFLD through the HDAC1/TAF9 pathway.
Collapse
Affiliation(s)
- Ruiwen Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Ruimin Sun
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Rong Fu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yu Sun
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Meiyang Zhu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yunfei Geng
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Dongyan Gao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Xue Z, Li Y, Zhou M, Liu Z, Fan G, Wang X, Zhu Y, Yang J. Traditional Herbal Medicine Discovery for the Treatment and Prevention of Pulmonary Arterial Hypertension. Front Pharmacol 2021; 12:720873. [PMID: 34899290 PMCID: PMC8660120 DOI: 10.3389/fphar.2021.720873] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary artery remodeling that may subsequently culminate in right heart failure and premature death. Although there are currently both non-pharmacological (lung transplantation, etc.) and pharmacological (Sildenafil, Bosentan, and new oral drugs on trial) therapies available, PAH remains a serious and fatal pulmonary disease. As a unique medical treatment, traditional herbal medicine (THM) treatment has gradually exerted its advantages in treating PAH worldwide through a multi-level and multi-target approach. Additionally, the potential mechanisms of THM were deciphered, including suppression of proliferation and apoptosis of pulmonary artery smooth muscle cells, controlling the processes of inflammation and oxidative stress, and regulating vasoconstriction and ion channels. In this review, the effects and mechanisms of the frequently studied compound THM, single herbal preparations, and multiple active components from THM are comprehensively summarized, as well as their related mechanisms on several classical preclinical PAH models. It is worth mentioning that sodium tanshinone IIA sulfonate sodium and tetramethylpyrazine are under clinical trials and are considered the most promoting medicines for PAH treatment. Last, reverse pharmacology, a strategy to discover THM or THM-derived components, has also been proposed here for PAH. This review discusses the current state of THM, their working mechanisms against PAH, and prospects of reverse pharmacology, which are expected to facilitate the natural anti-PAH medicine discovery and development and its bench-to-bedside transformation.
Collapse
Affiliation(s)
- Zhifeng Xue
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Yixuan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Mengen Zhou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Zhidong Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Xiaoying Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Jian Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| |
Collapse
|
19
|
Ding H, Yang X, Tian J, Wang X, Ji Y, El-Ashram S, Ren C, Shen J, Liu M. JQ-1 ameliorates schistosomiasis liver fibrosis by suppressing JAK2 and STAT3 activation. Biomed Pharmacother 2021; 144:112281. [PMID: 34624676 DOI: 10.1016/j.biopha.2021.112281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 01/09/2023] Open
Abstract
Schistosomiasis is a serious parasitic infection caused by Schistosoma. The parasite deposits eggs in the host liver, causing inflammation that activates hepatic stellate cells (HSCs), which leads to liver fibrosis. Currently, there is no effective therapy for liver fibrosis; thus, treatments are urgently needed. Therefore, in the present study, mice infected with Schistosoma japonicum were treated with JQ-1, a small-molecule bromodomain inhibitor with reliable anti-tumor and anti-inflammatory activities. The fibrotic area of the liver measured by computer-assisted morphometric analysis and the expression levels of the cytoskeletal protein alpha smooth muscle actin (α-SMA) and of collagen assessed by quantitative PCR, Western blot and immunohistochemistry were significantly decreased in the liver following JQ-1 treatment compared with vehicle-treated controls. Total RNA was extracted from the liver of JQ-1-treated Schistosoma-infected mice for RNA-sequencing analysis. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses indicated that JQ-1 affected biological processes and the expression of cellular components known to play key roles in the transdifferentiation of HSCs to myofibroblasts. In vitro treatment with JQ-1 of JS-1 cells, a mouse HSC line, indicated that JQ-1 significantly inhibited JS-1 proliferation but had no effect on JS-1 activity, senescence, or apoptosis. Western blot results showed that JQ-1 inhibited the expression levels of phosphorylated JAK2 and phosphorylated STAT3 without altering expression levels of these non-phosphorylated proteins. Taken together, these findings suggested that JQ-1 treatment ameliorated S. japonicum egg-induced liver fibrosis, at least in part, by suppressing HSC activation and proliferation through the inhibition of JAK2/STAT3 signaling. These results lay a foundation for the development of novel approaches to treat and control liver fibrosis caused by S. japonicum.
Collapse
Affiliation(s)
- Han Ding
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Xuhan Yang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Jiaming Tian
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Xinran Wang
- The Second Clinical Medical College, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Yongsheng Ji
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Saeed El-Ashram
- College of Life Science and Engineering, Foshan University, 18 Jiangwan Street, Foshan, Guangdong 528231 People's Republic of China
| | - Cuiping Ren
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China
| | - Jijia Shen
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China.
| | - Miao Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, People's Republic of China.
| |
Collapse
|
20
|
Xu L, Ling J, Su C, Su YW, Xu Y, Jiang Z. Emerging Roles on Immunological Effect of Indoleamine 2,3-Dioxygenase in Liver Injuries. Front Med (Lausanne) 2021; 8:756435. [PMID: 34869457 PMCID: PMC8636938 DOI: 10.3389/fmed.2021.756435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) is one of the initial rate-limiting enzymes of the kynurenine pathway (KP), which causes immune suppression and induction of T cell anergy. It is associated with the imbalance of immune homeostasis in numerous diseases including cancer, chronic viral infection, allergy, and autoimmune diseases. Recently, IDO has extended its role to liver field. In this review, we summarize the dysregulation and potentials of IDO in the emerging field of liver injuries, as well as current challenges for IDO targets. In particular, we discuss unexpected conclusions against previous work published. IDO is induced by pro-inflammatory cytokines in liver dysfunction and exerts an immunosuppressive effect, whereas the improvement of liver injury may require consideration of multiple factors besides IDO.
Collapse
Affiliation(s)
- Lingyan Xu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Jiawei Ling
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Chang Su
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Wen Su
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
21
|
Xu X, Hong P, Wang Z, Tang Z, Li K. MicroRNAs in Transforming Growth Factor-Beta Signaling Pathway Associated With Fibrosis Involving Different Systems of the Human Body. Front Mol Biosci 2021; 8:707461. [PMID: 34381815 PMCID: PMC8350386 DOI: 10.3389/fmolb.2021.707461] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis, a major cause of morbidity and mortality, is a histopathological manifestation of many chronic inflammatory diseases affecting different systems of the human body. Two types of transforming growth factor beta (TGF-β) signaling pathways regulate fibrosis: the canonical TGF-β signaling pathway, represented by SMAD-2 and SMAD-3, and the noncanonical pathway, which functions without SMAD-2/3 participation and currently includes TGF-β/mitogen-activated protein kinases, TGF-β/SMAD-1/5, TGF-β/phosphatidylinositol-3-kinase/Akt, TGF-β/Janus kinase/signal transducer and activator of transcription protein-3, and TGF-β/rho-associated coiled-coil containing kinase signaling pathways. MicroRNA (miRNA), a type of non-coding single-stranded small RNA, comprises approximately 22 nucleotides encoded by endogenous genes, which can regulate physiological and pathological processes in fibrotic diseases, particularly affecting organs such as the liver, the kidney, the lungs, and the heart. The aim of this review is to introduce the characteristics of the canonical and non-canonical TGF-β signaling pathways and to classify miRNAs with regulatory effects on these two pathways based on the influenced organ. Further, we aim to summarize the limitations of the current research of the mechanisms of fibrosis, provide insights into possible future research directions, and propose therapeutic options for fibrosis.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Pengyu Hong
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhefu Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Kun Li
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
22
|
Local Inhibition of Indoleamine 2,3-Dioxygenase Mitigates Renal Fibrosis. Biomedicines 2021; 9:biomedicines9080856. [PMID: 34440060 PMCID: PMC8389588 DOI: 10.3390/biomedicines9080856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern and renal fibrosis is an integral part of the pathophysiological mechanism underlying disease progression. In CKD patients, the majority of metabolic pathways are in disarray and perturbations in enzyme activity most likely contribute to the wide variety of comorbidities observed in these patients. To illustrate, catabolism of tryptophan by indoleamine 2,3-dioxygenase (IDO) gives rise to numerous biologically active metabolites implicated in CKD progression. Here, we evaluated the effect of antagonizing IDO on renal fibrogenesis. To this end, we antagonized IDO using 1-methyl-D-tryptophan (1-MT) and BMS-98620 in TGF-β-treated murine precision-cut kidney slices (mPCKS) and in mice subjected to unilateral ureteral obstruction (UUO). The fibrotic response was evaluated on both the gene and protein level using qPCR and western blotting. Our results demonstrated that treatment with 1-MT or BMS-985205 markedly reduced TGF-β-mediated fibrosis in mPCKS, as seen by a decreased expression of collagen type 1, fibronectin, and α-smooth muscle actin. Moreover, IDO protein expression clearly increased following UUO, however, treatment of UUO mice with either 1-MT or BMS-986205 did not significantly affect the gene and protein expression of the tested fibrosis markers. However, both inhibitors significantly reduced the renal deposition of collagen in UUO mice as shown by Sirius red and trichrome staining. In conclusion, this study demonstrates that IDO antagonism effectively mitigates fibrogenesis in mPCKS and reduces renal collagen accumulation in UUO mice. These findings warrant further research into the clinical application of IDO inhibitors for the treatment of renal fibrosis.
Collapse
|
23
|
Xu Z, Ke T, Zhang Y, Guo L, Chen F, He W. Danshensu inhibits the IL-1β-induced inflammatory response in chondrocytes and osteoarthritis possibly via suppressing NF-κB signaling pathway. Mol Med 2021; 27:80. [PMID: 34284715 PMCID: PMC8290616 DOI: 10.1186/s10020-021-00329-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Osteoarthritis (OA) is the most common inflammatory disease associated with pain and cartilage destruction. Interleukin (IL)-1β is widely used to induce inflammatory response in OA models. This study aimed to explore the role of Danshensu (DSS) in IL-1β-induced inflammatory responses in OA. Methods IL-1β was used to induce chondrocyte inflammation. Cell viability was evaluated by Cell Counting Kit-8 (CCK-8) assay. IL-6, COX-2, TNF-α, and iNOS mRNA levels were detected by qRT-PCR. MMP3, MMP13, ADAMTS4, ADAMTS5, Aggrecan, Collagen, p-IκBα, and p-p65 protein levels were detected by Western blot. An OA mouse model was established by surgical destabilization of the medial meniscus (DMM), and the Osteoarthritis Research Society International (OARSI) score was evaluated by H&E staining. Results DSS did not affect the levels of inflammatory indicators including IL-6, COX-2, TNF-α, iNOS, PEG2, and NO but suppressed COX-2 and iNOS protein expression in IL-1β treated chondrocytes. In addition, DSS downregulated IL-1β-enhanced expression of MMP3, MMP13, ADAMTS4, and ADAMTS5 and upregulated aggrecan and collagen expression. Moreover, DSS significantly inhibited IL-1β-induced phosphorylation of p-IκBα and p-p65 in a dose-dependent manner in chondrocytes, suggesting it plays a role in the NF-κB signaling pathway. Furthermore, DSS significantly reduced DMM-induced cartilage OARSI score in mice, further demonstrating its protective role in OA progression in vivo. Conclusions Our study revealed the protective role of DSS in OA, suggesting that DSS might act as a potential treatment for OA.
Collapse
Affiliation(s)
- Zhixian Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China.,Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Hospital, Fuzhou, 350001, Fujian, People's Republic of China
| | - Tie Ke
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Yongfa Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Licheng Guo
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China.,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Feng Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China. .,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China. .,Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Hospital, Fuzhou, 350001, Fujian, People's Republic of China.
| | - Wubing He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350004, Fujian Province, People's Republic of China. .,Department of Emergency Surgery, Fujian Provincial Hospital, No.134 East Street, Gulou District, Fuzhou, 350001, Fujian Province, People's Republic of China. .,Fujian Provincial Key Laboratory of Emergency Medicine, Fujian Provincial Hospital, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
24
|
Liu H, Zhang X, Shao Y, Lin X, Dong F, Liu X. Danshensu alleviates bleomycin-induced pulmonary fibrosis by inhibiting lung fibroblast-to-myofibroblast transition via the MEK/ERK signaling pathway. Bioengineered 2021; 12:3113-3124. [PMID: 34187349 PMCID: PMC8806824 DOI: 10.1080/21655979.2021.1944020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary fibrosis (PF) is a chronic pulmonary interstitial disease, and its pathological process is closely related to fibroblast-myofibroblast differentiation. Danshensu (DSS) has been reported to exert an anti-fibrotic effect in heart and liver. However, it is unknown whether DSS has an equally anti-fibrotic effect on lungs. To evaluate the effect of DSS on PF and demonstrate its possible molecular mechanisms, we established an in vitro model on TGF-β1 (5 ng/mL)-stimulated NIH3T3 cells and in vivo model on bleomycin (BLM) (5 mg/kg)-induced PF mice. In vitro, our results revealed that 50 μM DSS effectively inhibited the fibroblast proliferation, migration and differentiation into myofibroblast. In vivo, our results showed that DSS (28 and 56 mg/kg) reduced damaged lung structures, infiltrated inflammatory cells and accumulated areas of collagen deposition. Moreover, we showed that DSS decreased the fibroblast-specific protein 1 (FSP-1) - and α-SMA-positive areas. Meanwhile, we indicated that DSS reduced the expression of TGF-β1, α-SMA and COL-I in the lung tissues of mice. To further explore the mechanism of DSS on alleviating PF, we detected the MEK/ERK signaling pathway. Our results showed that DSS reduced the phosphorylation of MEK1/2 and ERK1/2, indicating that DSS might inhibit the MEK/ERK signaling pathway. Taken together, these results demonstrated that DSS could suppress lung fibroblast proliferation, migration and differentiation to myofibroblasts, possibly through suppressing the MEK/ERK signaling pathway, which suggested that DSS might be a potential therapeutic drug for PF treatment.
Collapse
Affiliation(s)
- Huaman Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of General Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyue Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yumeng Shao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuehong Lin
- Department of General Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feng Dong
- Department of General Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Respiration, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
25
|
Zhao X, Zhao B, Zhao Y, Zhang Y, Qian M. Protective effect of anisodamine on bleomycin-induced acute lung injury in immature rats via modulating oxidative stress, inflammation, and cell apoptosis by inhibiting the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:859. [PMID: 34164493 PMCID: PMC8184412 DOI: 10.21037/atm-21-1750] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Pediatric acute lung injury (ALI) is one of the most common causes of infant mortality. Although lung-protective strategies have developed in recent years, no ALI treatment is currently available. Anisodamine (Ani) is a common drug used to treat gastrointestinal smooth muscle spasm. The protective effects of Ani against acute kidney injury and myocardial injury have been reported. However, the efficacy of Ani on bleomycin (BLM)-induced ALI has not been examined previously. In the present study, we aimed to examine the effects of Ani on bleomycin (BLM)-induced ALI on immature rats. Methods The ALI rat model was established by intratracheally administration of BLM. Ani treatment was performed by an intravenous injection at different concentrations. The lung function of each rat was measured, and then lung tissue structures, apoptosis, and collagen deposition were observed by hematoxylin-eosin staining, terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assay, and Masson’s staining, respectively. Enzyme-linked immunosorbent assay was used to detect the levels of inflammatory cytokines. The expression of apoptosis-related proteins and fibrosis-related markers was determined by reverse transcription-polymerase chain reaction and/or Western blot analysis. Finally, the expression levels of Janus tyrosine kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were determined. Results Our findings indicated that lung function was remarkably decreased in BLM-induced rats, which could be reversed by Ani. Ani treatment increased the levels of antioxidant enzymes, decreased the apoptotic rate and apoptosis-related proteins, and downregulated the expression of fibrosis-related markers. Additionally, Ani treatment also attenuated inflammatory response and suppressed the activation of the JAK2/STAT3 pathway. Conclusions Our results demonstrated that Ani had potent activity against BLM-induced ALI in immature rats through inhibiting the JAK2/STAT3 signaling pathway. Our findings provide supporting evidence to further investigate the therapeutic effect of Ani against ALI in children.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Bin Zhao
- Department of Neurosurgery, the Second Hospital of Jilin University, Changchun, China
| | - Yinghao Zhao
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Yunfeng Zhang
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Min Qian
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Mo C, Xie S, Zeng T, Lai Y, Huang S, Zhou C, Yan W, Huang S, Gao L, Lv Z. Ginsenoside-Rg1 acts as an IDO1 inhibitor, protects against liver fibrosis via alleviating IDO1-mediated the inhibition of DCs maturation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153524. [PMID: 33667840 DOI: 10.1016/j.phymed.2021.153524] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/19/2021] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase 1 (IDO1) has been reported as a hallmark of hepatic fibrosis. Ginseng Rg1(G-Rg1) is a characterized bioactive component isolated from a traditional Chinese medicinal herb Panax ginseng C. A. Meyer (Ginseng) that used in China widely. However, the anti-hepatic fibrosis property of G-Rg1 and the underlying mechanisms of action are poorly reported. PURPOSE Here, we researched the effect of G-Rg1 on experimental liver fibrosis in vivo and in vitro. STUDY DESIGN AND METHODS We applied a CCL4-induced liver fibrosis in mice (wild-type and those overexpressing IDO1 by in vivo AAV9 vector) and HSC-T6 cells to detect the anti-hepatic fibrosis effect of G-Rg1 in vivo and in vitro. RESULTS We found that G-Rg1 reduced serum levels of AST and ALT markedly. Histologic examination indicated that G-Rg1 dramatically improved the extent of liver fibrosis and suppressed the hepatic levels of fibrotic marker α-SMA in vivo and in vitro. The proliferation of HSC-T6 was significantly inhibited by G-Rg1 in vitro. Both TUNEL staining and flow cytometry demonstrated that G-Rg1 attenuated the levels of hepatocyte apoptosis in fibrotic mice. Additionally, G-Rg1 up-regulated the maturation of hepatic DCs via reducing the expression level of hepatic IDO1, which played an inverse role in the maturation of DCs. Furthermore, oral administration of G-Rg1 ameliorated IDO1 overexpression-induced worsen liver fibrosis as well as IDO1 overexpression-mediated more apparent inhibition of maturation of DCs. CONCLUSION These results suggest that G-Rg1, which exerts its antifibrotic properties via alleviating IDO1-mediated the inhibition of DCs maturation, may be a potential therapeutic drug in treating liver fibrosis.
Collapse
Affiliation(s)
- Chan Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Weixin Yan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Shaohui Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China; The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou510515, PR China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China.
| |
Collapse
|
27
|
Xiao G, Lyu M, Li Z, Cao L, Liu X, Wang Y, He S, Chen Z, Du H, Feng Y, Wang J, Zhu Y. Restoration of early deficiency of axonal guidance signaling by guanxinning injection as a novel therapeutic option for acute ischemic stroke. Pharmacol Res 2021; 165:105460. [PMID: 33513357 DOI: 10.1016/j.phrs.2021.105460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 01/12/2023]
Abstract
Despite of its high morbidity and mortality, there is still a lack of effective treatment for ischemic stroke in part due to our incomplete understanding of molecular mechanisms of its pathogenesis. In this study, we demonstrate that SHH-PTCH1-GLI1-mediated axonal guidance signaling and its related neurogenesis, a central pathway for neuronal development, also plays a critical role in early stage of an acute stroke model. Specifically, in vivo, we evaluated the effect of GXNI on ischemic stroke mice via using the middle cerebral artery embolization model, and found that GXNI significantly alleviated cerebral ischemic reperfusion (I/R) injury by reducing the volume of cerebral infarction, neurological deficit score and cerebral edema, reversing the BBB permeability and histopathological changes. A combined approach of RNA-seq and network pharmacology analysis was used to reveal the underlying mechanisms of GXNI followed by RT-PCR, immunohistochemistry and western blotting validation. It was pointed out that axon guidance signaling pathway played the most prominent role in GXNI action with Shh, Ptch1, and Gli1 genes as the critical contributors in brain protection. In addition, GXNI markedly prevented primary cortical neuron cells from oxygen-glucose deprivation/reoxygenation damage in vitro, and promoted axon growth and synaptogenesis of damaged neurons, which further confirmed the results of in vivo experiments. Moreover, due to the inhibition of the SHH-PTCH1-GLI1 signaling pathway by cyclopropylamine, the effect of GXNI was significantly weakened. Hence, our study provides a novel option for the clinical treatment of acute ischemic stroke by GXNI via SHH-PTCH1-GLI1-mediated axonal guidance signaling, a neuronal development pathway previously considered for after-stroke recovery.
Collapse
Affiliation(s)
- Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Linghua Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Zihao Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Jigang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
28
|
Mo C, Xie S, Liu B, Zhong W, Zeng T, Huang S, Lai Y, Deng G, Zhou C, Yan W, Chen Y, Huang S, Gao L, Lv Z. Indoleamine 2,3-dioxygenase 1 limits hepatic inflammatory cells recruitment and promotes bile duct ligation-induced liver fibrosis. Cell Death Dis 2021; 12:16. [PMID: 33414436 PMCID: PMC7791029 DOI: 10.1038/s41419-020-03277-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is a course of chronic liver dysfunction, can develop into cirrhosis and hepatocellular carcinoma. Inflammatory insult owing to pathogenic factors plays a crucial role in the pathogenesis of liver fibrosis. Indoleamine 2,3-dioxygenase 1 (IDO1) can affect the infiltration of immune cells in many pathology processes of diseases, but its role in liver fibrosis has not been elucidated completely. Here, the markedly elevated protein IDO1 in livers was identified, and dendritic cells (DCs) immune-phenotypes were significantly altered after BDL challenge. A distinct hepatic population of CD11c+DCs was decreased and presented an immature immune-phenotype, reflected by lower expression levels of co-stimulatory molecules (CD40, MHCII). Frequencies of CD11c+CD80+, CD11c+CD86+, CD11c+MHCII+, and CD11c+CD40+ cells in splenic leukocytes were reduced significantly. Notably, IDO1 overexpression inhibited hepatic, splenic CD11c+DCs maturation, mature DCs-mediated T-cell proliferation and worsened liver fibrosis, whereas above pathological phenomena were reversed in IDO1-/- mice. Our data demonstrate that IDO1 affects the process of immune cells recruitment via inhibiting DCs maturation and subsequent T cells proliferation, resulting in the promotion of hepatic fibrosis. Thus, amelioration of immune responses in hepatic and splenic microenvironment by targeting IDO1 might be essential for the therapeutic effects on liver fibrosis.
Collapse
Affiliation(s)
- Chan Mo
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Bin Liu
- Department of Emergency, Guangzhou Red Cross Hospital, Medical College, Jinan University, 510220, Guangzhou, China
| | - Weichao Zhong
- Shenzhen Traditional Chinese Medicine Hospital, No.1, Fuhua Road, Futian District, 518033, Shenzhen, Guangdong, People's Republic of China
| | - Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Weixin Yan
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Shaohui Huang
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, 510515, Guangzhou, People's Republic of China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
29
|
Wang RN, Zhao HC, Huang JY, Wang HL, Li JS, Lu Y, Di LQ. Challenges and strategies in progress of drug delivery system for traditional Chinese medicine Salviae Miltiorrhizae Radix et Rhizoma (Danshen). CHINESE HERBAL MEDICINES 2021; 13:78-89. [PMID: 36117766 PMCID: PMC9476708 DOI: 10.1016/j.chmed.2020.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/14/2020] [Accepted: 08/18/2020] [Indexed: 12/29/2022] Open
Abstract
Traditional Chinese medicines (TCMs), with a history of thousands of years, are widely used clinically with effective treatment. However, the drug delivery systems (DDSs) for TCMs remains major challenges due to the characteristics of multi-components including alkaloids, flavones, anthraquinones, glycosides, proteins, volatile oils and other types. Therefore, the novel preparations and technology of modern pharmaceutics is introduced to improve TCM therapeutic effects due to instability and low bioavailability of active ingredients. Salviae Miltiorrhizae Radix et Rhizoma, the radix and rhizomes of Salvia miltiorrhiza Bunge (Danshen in Chinese), is a well known Chinese herbal medicine for protecting the cardiovascular system, with active ingredients mainly including lipophilic tanshinones and hydrophilic salvianolic acids. In this review, this drug is taken as an example to present challenges and strategies in progress of DDSs for TCMs. This review would also summary the characteristics of active ingredients in it including physicochemical properties and pharmacological effects. The purpose of this review is to provide inspirations and ideas for the DDSs designed from TCMs by summarizing the advances on DDSs for both single- and multi-component from Danshen.
Collapse
Affiliation(s)
- Ruo-ning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
- Corresponding authors.
| | - Hua-cong Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
| | - Jian-yu Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
| | - Hong-lan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
| | - Jun-song Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
| | - Yin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing 210046, China
| | - Liu-qing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing 210046, China
- Corresponding authors.
| |
Collapse
|
30
|
Zhang Y, Zhang G, Liang Y, Wang H, Wang Q, Zhang Y, Zhang X, Zhang J, Chu L. Potential Mechanisms Underlying the Hepatic–Protective Effects of Danshensu on Iron Overload Mice. Biol Pharm Bull 2020; 43:968-975. [DOI: 10.1248/bpb.b19-01084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Hebei University of Chinese Medicine
| | - Gaohua Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine
| | - Yingran Liang
- School of Basic Medicine, Hebei University of Chinese Medicine
| | - Hongfang Wang
- School of Pharmacy, Hebei University of Chinese Medicine
| | - Qian Wang
- School of Pharmacy, Hebei University of Chinese Medicine
| | - Ying Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine
| | - Xuan Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine
| | - Jianping Zhang
- School of Basic Medicine, Hebei University of Chinese Medicine
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns
| |
Collapse
|