101
|
Saha P, Talukdar AD, Nath R, Sarker SD, Nahar L, Sahu J, Choudhury MD. Role of Natural Phenolics in Hepatoprotection: A Mechanistic Review and Analysis of Regulatory Network of Associated Genes. Front Pharmacol 2019; 10:509. [PMID: 31178720 PMCID: PMC6543890 DOI: 10.3389/fphar.2019.00509] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
The liver is not only involved in metabolism and detoxification, but also participate in innate immune function and thus exposed to frequent target Thus, they are the frequent target of physical injury. Interestingly, liver has the unique ability to regenerate and completely recoup from most acute, non-iterative situation. However, multiple conditions, including viral hepatitis, non-alcoholic fatty liver disease, long term alcohol abuse and chronic use of medications can cause persistent injury in which regenerative capacity eventually becomes dysfunctional resulting in hepatic scaring and cirrhosis. Despite the recent therapeutic advances and significant development of modern medicine, hepatic diseases remain a health problem worldwide. Thus, the search for the new therapeutic agents to treat liver disease is still in demand. Many synthetic drugs have been demonstrated to be strong radical scavengers, but they are also carcinogenic and cause liver damage. Present day various hepatic problems are encountered with number of synthetic and plant based drugs. Nexavar (sorafenib) is a chemotherapeutic medication used to treat advanced renal cell carcinoma associated with several side effects. There are a few effective varieties of herbal preparation like Liv-52, silymarin and Stronger neomin phages (SNMC) against hepatic complications. Plants are the huge repository of bioactive secondary metabolites viz; phenol, flavonoid, alkaloid etc. In this review we will try to present exclusive study on phenolics with its mode of action mitigating liver associated complications. And also its future prospects as new drug lead.
Collapse
Affiliation(s)
- Priyanka Saha
- Department of Life Science & Bioinformatics, Assam University, Silchar, India
| | - Anupam Das Talukdar
- Department of Life Science & Bioinformatics, Assam University, Silchar, India
| | - Rajat Nath
- Department of Life Science & Bioinformatics, Assam University, Silchar, India
| | - Satyajit D. Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Lutfun Nahar
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jagajjit Sahu
- Department of Mycology and Plant Pathology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
102
|
Lee WP, Lan KL, Liao SX, Huang YH, Hou MC, Lan KH. Antiviral effect of saikosaponin B2 in combination with daclatasvir on NS5A resistance-associated substitutions of hepatitis C virus. J Chin Med Assoc 2019; 82:368-374. [PMID: 30920421 DOI: 10.1097/jcma.0000000000000095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Hepatitis C virus (HCV) is a major causative agent of chronic hepatitis, cirrhosis, and hepatocellular carcinoma. The rapid progress in the development of direct-acting antivirals has greatly elevated the cure rate to ≥95% in recent years. However, the high cost of treatment is not affordable to patients in some countries, necessitating the development of less expensive treatment. METHODS We adopted a cell culture-derived HCV system to screen a library of the pure compounds extracted from herbs deposited in the chemical bank of the National Research Institute of Chinese Medicine, Taiwan. RESULTS We found that saikosaponin B2 inhibited viral entry, replication, and translation. Saikosaponin B2 is a plant glycoside and a component of xiao-chai-hu-tang, a traditional Chinese herbal medicine extracted from the roots of Bupleurum falcatum. It also inhibited daclatasvir-resistant mutant strains of HCV, especially in combination with daclatasvir. CONCLUSION Our results may aid the development of a new combination therapy useful for patients with HCV who are intolerant or refractory to the currently available medications, including pegylated interferon and direct-acting antiviral agents.
Collapse
Affiliation(s)
- Wei-Ping Lee
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Keng-Li Lan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Shi-Xian Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Keng-Hsin Lan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
103
|
Zhang T, Kawaguchi N, Yoshihara K, Hayama E, Furutani Y, Kawaguchi K, Tanaka T, Nakanishi T. Silibinin efficacy in a rat model of pulmonary arterial hypertension using monocrotaline and chronic hypoxia. Respir Res 2019; 20:79. [PMID: 31023308 PMCID: PMC6485095 DOI: 10.1186/s12931-019-1041-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 04/02/2019] [Indexed: 01/07/2023] Open
Abstract
Background C-X-C chemokine receptor type 4 (CXCR4) may be involved in the development of pulmonary arterial hypertension (PAH). CXCR4 inhibitor AMD3100 was described to have a positive effect on the prevention of pulmonary arterial muscularization in PAH models. Silibinin is a traditional medicine that has an antagonistic effect on CXCR4. We investigated the effect of silibinin using rat models of PAH. Methods PAH was induced by a single subcutaneous injection of monocrotaline. The rats were maintained in a chronic hypoxic condition (10% O2) with or without silibinin. To evaluate the efficacy of silibinin on PAH, right ventricular systolic pressure (RVSP), Fulton index (weight ratio of right ventricle to the left ventricle and septum), percent medial wall thickness (% MT), and vascular occlusion score (VOS) were measured and calculated. Immunohistochemical analysis was performed targeting CXCR4 and c-Kit. Reverse transcription-quantitative polymerase chain reaction was performed for the stem cell markers CXCR4, stromal cell derived factor-1 (SDF-1), c-Kit, and stem cell factor (SCF), and the inflammatory markers monocyte chemoattractant protein 1 (MCP1), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNFα). Statistical analyses were performed using t-test and one-way analysis of variance with Bonferroni’s post hoc test. Results Silibinin treatment for 1 week reduced RVSP and Fulton index. Treatment for 2 weeks reduced RVSP, Fulton index, % MT, and VOS, as well as downregulating the expression of CXCR4, SDF-1, and TNFα in pulmonary arteries. In contrast, treatment for 3 weeks failed to ameliorate PAH. The time-course study demonstrated that RVSP, Fulton index, % MT, and VOS gradually increased over time, with a decrease in the expression of CXCR4 and TNFα occurring after 2 weeks of PAH development. After 3 weeks, SDF-1, c-Kit, and SCF began to decrease and, after 5 weeks, MCP1 and IL-6 gradually accumulated. Conclusions The CXCR4 inhibitor silibinin can ameliorate PAH, possibly through the suppression of the CXCR4/SDF-1 axis, until the point where PAH becomes a severe and irreversible condition. Silibinin results in reduced pulmonary arterial pressure and delays pulmonary arteriolar occlusion and pulmonary vascular remodeling. Electronic supplementary material The online version of this article (10.1186/s12931-019-1041-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan.,Department of Structural Heart Disease, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Nanako Kawaguchi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan.
| | - Kenji Yoshihara
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Emiko Hayama
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Yoshiyuki Furutani
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Kayoko Kawaguchi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Takeshi Tanaka
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Toshio Nakanishi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan.
| |
Collapse
|
104
|
The Edible Insect Gryllus bimaculatus Protects against Gut-Derived Inflammatory Responses and Liver Damage in Mice after Acute Alcohol Exposure. Nutrients 2019; 11:nu11040857. [PMID: 30995745 PMCID: PMC6521266 DOI: 10.3390/nu11040857] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/10/2019] [Accepted: 04/13/2019] [Indexed: 12/12/2022] Open
Abstract
Accumulation of reactive oxygen species (ROS) in response to excess alcohol exposure is a major cause of gut barrier disruption and lipopolysaccharide (LPS)-induced hepatic inflammation, as well as liver steatosis and apoptosis. This study was designed to investigate protective effects of the cricket Gryllus bimaculatus, an edible insect recognized by the Korea Food and Drug Administration, against acute alcoholic liver damage in mice. Administration of G. bimaculatus extracts (GBE) attenuated alcohol-induced steatosis and apoptotic responses in the liver and intestinal permeability to bacterial endotoxin. These protective effects were associated with suppression of ROS-mediated oxidative stress in both the liver and small intestine. Furthermore, in vivo and in vitro studies revealed that GBE inhibits LPS-induced Kupffer cell activation and subsequent inflammatory signaling. Importantly, the protective effects of GBE were more potent than those of silymarin, a known therapeutic agent for alcoholic liver diseases.
Collapse
|
105
|
Pérez-Sánchez A, Cuyàs E, Ruiz-Torres V, Agulló-Chazarra L, Verdura S, González-Álvarez I, Bermejo M, Joven J, Micol V, Bosch-Barrera J, Menendez JA. Intestinal Permeability Study of Clinically Relevant Formulations of Silibinin in Caco-2 Cell Monolayers. Int J Mol Sci 2019; 20:E1606. [PMID: 30935093 PMCID: PMC6480586 DOI: 10.3390/ijms20071606] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
An ever-growing number of preclinical studies have investigated the tumoricidal activity of the milk thistle flavonolignan silibinin. The clinical value of silibinin as a bona fide anti-cancer therapy, however, remains uncertain with respect to its bioavailability and blood⁻brain barrier (BBB) permeability. To shed some light on the absorption and bioavailability of silibinin, we utilized the Caco-2 cell monolayer model of human intestinal absorption to evaluate the permeation properties of three different formulations of silibinin: silibinin-meglumine, a water-soluble form of silibinin complexed with the amino-sugar meglumine; silibinin-phosphatidylcholine, the phytolipid delivery system Siliphos; and Eurosil85/Euromed, a milk thistle extract that is the active component of the nutraceutical Legasil with enhanced bioavailability. Our approach predicted differential mechanisms of transport and blood⁻brain barrier permeabilities between the silibinin formulations tested. Our assessment might provide valuable information about an idoneous silibinin formulation capable of reaching target cancer tissues and accounting for the observed clinical effects of silibinin, including a recently reported meaningful central nervous system activity against brain metastases.
Collapse
Affiliation(s)
- Almudena Pérez-Sánchez
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Verónica Ruiz-Torres
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Luz Agulló-Chazarra
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Isabel González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Marival Bermejo
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Vicente Micol
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), 07122 Palma de Mallorca, Spain.
| | - Joaquim Bosch-Barrera
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Department of Medical Sciences, Medical School University of Girona, 17003 Girona, Spain.
- Medical Oncology, Catalan Institute of Oncology (ICO), Dr. Josep Trueta University Hospital, 17007 Girona, Spain.
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| |
Collapse
|
106
|
Mohammadi H, Hadi A, Arab A, Moradi S, Rouhani MH. Effects of silymarin supplementation on blood lipids: A systematic review and meta‐analysis of clinical trials. Phytother Res 2019; 33:871-880. [DOI: 10.1002/ptr.6287] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/15/2018] [Accepted: 12/19/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Hamed Mohammadi
- Student Research Committee, Department of Clinical Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical Sciences Isfahan Iran
| | - Amir Hadi
- Halal Research Center of IRIFDA Tehran Iran
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research CenterIsfahan University of Medical Sciences Isfahan Iran
| | - Arman Arab
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research CenterIsfahan University of Medical Sciences Isfahan Iran
| | - Sajjad Moradi
- Halal Research Center of IRIFDA Tehran Iran
- Nutritional Sciences Department, School of Nutritional Sciences and Food TechnologyKermanshah University of Medical Sciences Kermanshah Iran
| | - Mohammad Hossein Rouhani
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research CenterIsfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
107
|
Theodoropoulou M, Reincke M. Tumor-Directed Therapeutic Targets in Cushing Disease. J Clin Endocrinol Metab 2019; 104:925-933. [PMID: 30535260 DOI: 10.1210/jc.2018-02080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
CONTEXT The most frequent cause of endogenous hypercortisolism is Cushing disease (CD), a devastating condition associated with severe comorbidities and high mortality. Effective tumor-targeting therapeutics are limited. DESIGN Search in PubMed with key words "corticotroph" and "Cushing's disease" plus the name of the mentioned therapeutic agent and in associated references of the obtained papers. Additionally, potential therapeutics were obtained from ClinicalTrials.gov with a search for "Cushing disease." RESULTS At present, the tumor-targeted pharmacological therapy of CD is concentrated on dopamine agonists (cabergoline) and somatostatin analogs (pasireotide) with varying efficacy, escape from response, and considerable side effects. Preclinical studies on corticotroph pathophysiology have brought forward potential drugs such as retinoic acid, silibinin, and roscovitine, whose efficacy and safety remain to be determined. CONCLUSIONS For many patients with CD, effective tumor-targeted pharmacological therapy is still lacking. Coordinated efforts are pivotal in establishing efficacy and safety of novel therapeutics in this rare but devastating disease.
Collapse
Affiliation(s)
- Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, Ludwig Maximilian University Munich, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Ludwig Maximilian University Munich, Munich, Germany
| |
Collapse
|
108
|
Nouri A, Heidarian E. Nephroprotective effect of silymarin against diclofenac induced renal damage and oxidative stress in male rats. JOURNAL OF HERBMED PHARMACOLOGY 2019. [DOI: 10.15171/jhp.2019.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction: Diclofenac (DIC), a phenylacetic acid compound which belongs to nonsteroidal anti-inflammatory drugs (NSAIDs), is generally used for the treatment of various diseases such as rheumatoid arthritis, ankylosing spondylitis, acute muscle pain conditions and osteoarthritis. Overdose of DIC can lead to renal injuries in both experimental animal and human. Our research was done to assess the protective role of silymarin on renal damage induced by DIC in rats. Methods: Thirty-two Wistar rats were assigned to four groups (n=8/group). Group 1 was control group; animals in group 2 were administrated DIC; Groups 3 and 4 administrated DIC plus silymarin with doses of 100 mg/kg and 200 mg/kg, orally (p.o), respectively. Various biochemical, molecular, and histological parameters were evaluated in serum and tissue homogenate. Results: In the second group, the levels of kidney catalase (CAT), vitamin C and superoxide dismutase (SOD) remarkably reduced (P < 0.05) relative to the control group. Also, urea, creatinine (Cr), malondialdehyde (MDA), serum tumor necrosis factor-α (TNF-α) and gene expression of TNF-α in this group were noticeably elevated (P < 0.05) relative to the control group. Treatment with silymarin caused a remarkable elevation (P < 0.05) in vitamin C, SOD, CAT and a remarkable reduction (P < 0.05) in the content of MDA, urea, Cr, TNF-α gene expression and serum TNF-α in comparison with second group. Histological injuries were also ameliorated by silymarin administration. Conclusion: The results confirm that silymarin has an ameliorative role against renal damage and oxidative stress induced by DIC in male rats.
Collapse
Affiliation(s)
- Ali Nouri
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Esfandiar Heidarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
109
|
Involvement of CXCR4 in Normal and Abnormal Development. Cells 2019; 8:cells8020185. [PMID: 30791675 PMCID: PMC6406665 DOI: 10.3390/cells8020185] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/30/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
CXC motif chemokine receptor type 4 (CXCR4) is associated with normal and abnormal development, including oncogenesis. The ligand of CXCR4 is stromal cell-derived factor (SDF), also known as CXC motif ligand (CXCL) 12. Through the SDF-1/CXCR4 axis, both homing and migration of hematopoietic (stem) cells are regulated through niches in the bone marrow. Outside of the bone marrow, however, SDF-1 can recruit CXCR4-positive cells from the bone marrow. SDF/CXCR4 has been implicated in the maintenance and/or differentiation of stemness, and tissue-derived stem cells can be associated with SDF-1 and CXCR4 activity. CXCR4 plays a role in multiple pathways involved in carcinogenesis and other pathologies. Here, we summarize reports detailing the functions of CXCR4. We address the molecular signature of CXCR4 and how this molecule and cells expressing it are involved in either normal (maintaining stemness or inducing differentiation) or abnormal (developing cancer and other pathologies) events. As a constituent of stem cells, the SDF-1/CXCR4 axis influences downstream signal transduction and the cell microenvironment.
Collapse
|
110
|
Grattagliano I, Montezinho LP, Oliveira PJ, Frühbeck G, Gómez-Ambrosi J, Montecucco F, Carbone F, Wieckowski MR, Wang DQH, Portincasa P. Targeting mitochondria to oppose the progression of nonalcoholic fatty liver disease. Biochem Pharmacol 2019; 160:34-45. [PMID: 30508523 DOI: 10.1016/j.bcp.2018.11.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a condition characterized by the excessive accumulation of triglycerides in hepatocytes. NAFLD is the most frequent chronic liver disease in developed countries, and is often associated with metabolic disorders such as obesity and type 2 diabetes. NAFLD definition encompasses a spectrum of chronic liver abnormalities, ranging from simple steatosis (NAFL), to steatohepatitis (NASH), significant liver fibrosis, cirrhosis, and hepatocellular carcinoma. NAFLD, therefore, represents a global public health issue. Mitochondrial dysfunction occurs in NAFLD, and contributes to the progression to the necro-inflammatory and fibrotic form (NASH). Disrupted mitochondrial function is associated with a decrease in the energy levels and impaired redox balance, and negatively affects cell survival by altering overall metabolism and subcellular trafficking. Such events reduce the tolerance of hepatocytes towards damaging hits, and favour the injurious effects of extra-cellular factors. Here, we discuss the role of mitochondria in NAFLD and focus on potential therapeutic approaches aimed at preserving mitochondrial function.
Collapse
Affiliation(s)
- Ignazio Grattagliano
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy; Italian College of General Practitioners and Primary Care, Bari, Italy
| | - Liliana P Montezinho
- CNC Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Center for Investigation Vasco da Gama (CIVG), Department of Veterinary Medicine, Escola Universitária Vasco da Gama, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Ospedale Policlinico San Martino, 10 Largo Benzi, 16132 Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 Viale Benedetto XV, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | | | - David Q-H Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
111
|
Nahum MS, Miguel DM, Jahir SN, Ramón SM, Flores-Murrieta J. Superior silybin bioavailability of silybin-phosphatidylcholine complex in oily-medium soft-gel capsules versus conventional silymarin tablets in healthy volunteers. BMC Pharmacol Toxicol 2019; 20:5. [PMID: 30635055 PMCID: PMC6330464 DOI: 10.1186/s40360-018-0280-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 12/13/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Fibrosis is a response to chronic liver disease that results in excessive accumulation of extracellular matrix proteins and formation of scar tissue. Fibrosis represents a clinical challenge of worldwide significance. Several studies have demonstrated that many natural products and herbal medicines have activity against liver fibrosis, and extracts of milk thistle such as silymarin and silybin are the natural compounds most commonly prescribed for liver diseases. Therefore, we sought to assess and compare the pharmacokinetic properties and bioavailability of silybin-phosphatidylcholine complex in oily-medium soft-gel capsules and conventional silymarin tablets in healthy Mexican volunteers. METHODS We enrolled 23 healthy volunteers to participate in a prospective, balanced, blind, single-dose, two-way crossover study with a one-week washout period. Fasting participants received either 45 mg silybin-phosphatidylcholine complex or 70 mg silymarin to assess which formulation provided better bioavailability of silybin. Plasma was obtained and analysed for silybin concentration using a validated ultra-performance liquid chromatography-tandem mass spectroscopy method. Pharmacokinetic parameters were obtained by non-compartmental analysis and values were compared by analysis of variance for a crossover design. Ratios of maximum plasma drug concentration and area under the curve (AUC) were obtained and 90% confidence intervals were calculated. RESULTS The 23 healthy subjects (11 women, 12 men) who participated in the study were aged 22-31 years old (average: 28), average weight 64.8 kg, height 1.65 m and body mass index 23.5 kg/m2. Plasma levels of silybin were higher after the administration of silybin-phosphatidylcholine complex capsules compared with that after conventional silymarin tablets (P < 0.0001). CONCLUSIONS The silybin-phosphatidylcholine complex in oily-medium soft-gel capsules seems to provide superior bioavailability. However, clinical studies must be performed to demonstrate its clinical relevance in the treatment of liver diseases. TRIAL REGISTRATION NCT03440164 ; registered on November 11, 2016.
Collapse
Affiliation(s)
- Méndez-Sánchez Nahum
- Liver Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Col. Toriello Guerra, 14050 Mexico City, Mexico
| | | | | | | | - J.Francisco Flores-Murrieta
- National Institute of Respiratory Diseases “Ismael Cosío Villegas”, 14080 Mexico City, Mexico
- Superior School of Medicine, National Polytechnic Institute of Mexico, 14080 Mexico City, Mexico
| |
Collapse
|
112
|
Tavassoli M, Afshari A, Arsene AL, Mégarbane B, Dumanov J, Paoliello MMB, Tsatsakis A, Carvalho F, Hashemzaei M, Karimi G, Rezaee R. Toxicological profile of Amanita virosa - A narrative review. Toxicol Rep 2019; 6:143-150. [PMID: 30705830 PMCID: PMC6348736 DOI: 10.1016/j.toxrep.2019.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/08/2019] [Indexed: 01/06/2023] Open
Abstract
Mushrooms account for a part of human diet due to their exquisite taste and protein content as well as their promising health effects unveiled by scientific research. Toxic and non-toxic mushrooms frequently share considerable morphological similarities, which mislead the collectors/consumers, resulting in mycotoxicity. Numerous mushroom species are considered "poisonous" as they produce dangerous toxins. For instance, members of the genus Amanita, especially A. phalloides, A. virosa and A. verna, are responsible for severe and even life-threatening noxious consequences. Globally, mushroom poisoning is a crucial healthcare issue as it leads to a considerable number of deaths annually. However, no definite antidote has been introduced to treat this poisoning. The present article discusses the characteristics of A. virosa in terms of epidemiology, mechanisms of toxicity, poisoning features and management.
Collapse
Affiliation(s)
- Milad Tavassoli
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Afshari
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Andree Letiţia Arsene
- Department of General and Pharmaceutical Microbiology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956, Bucharest, Romania
| | - Bruno Mégarbane
- Department of Medical and Toxicological Critical Care, Paris-Diderot University, INSERM UMRS-1144, Paris, France
| | - Josef Dumanov
- Mycological Institute USA EU, SubClinical Research Group, Sparta, NJ 07871, United States
| | - Monica Maria Bastos Paoliello
- Graduate Program in Public Health, Center of Health Sciences, State University of Londrina – UEL, Londrina, Paraná, Brazil
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
113
|
Abstract
Alcohol consumption is one of the main risks to public health. Alcohol use disorders (AUDs) cause 80% of hepatotoxic deaths, and approximately 50% of cirrhosis is alcohol-related. The acceptable daily intake (ADI) for ethanol is 2.6 g/day, deduced from morbidity and mortality rates due to liver fibrosis. The relative risk of cirrhosis increases significantly for doses above 60 g/day for men and 20 g/day for women over a period of around 10 years. Twenty to 40% of steatosis cases will evolve into steatohepatitis/steatofibrosis, and 8 to 20% will evolve directly into liver cirrhosis. About 20 to 40% of steatohepatitis cases will evolve into cirrhosis, and 4 to 5% into hepatocellular carcinoma. This cascade of events evolves in 5 to 40 years, with the temporal variability caused by the subjects' genetic patterns and associated risk/comorbidity factors. Steatohepatitis should be considered "the rate limiting step:" usually, it can be resolved through abstinence, although for some patients, once this situation develops, it is not substantially modified by abstention and there is a risk of fibrotic evolution. Early detection of fibrosis, obtained by hepatic elastography, is a crucial step in patients with AUDs. Such strategy allows patients to be included in a detoxification program in order to achieve abstention. Drugs such as silybin, metadoxine, and adenosylmethionine can be used. Other drugs, with promising antifibrotic effects, are currently under study. In this review, we discuss clinical and pathogenetic aspects of alcohol-related liver fibrosis and present and future strategies to prevent cirrhosis.
Collapse
Affiliation(s)
- Gianni Testino
- Alcohological Regional Center, Ligurian Region, ASL3, San Martino Hospital, Genoa, Italy -
| | - Silvia Leone
- Alcohological Regional Center, Ligurian Region, ASL3, San Martino Hospital, Genoa, Italy
| | - Sharmila Fagoonee
- Institute for Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | | |
Collapse
|
114
|
Kuo YJ, Chang HP, Chang YJ, Wu HH, Chen CH. Evaluation of nephroprotection of silymarin on contrast-induced nephropathy in liver cirrhosis patients: A population-based cohort study. Medicine (Baltimore) 2018; 97:e12243. [PMID: 30212956 PMCID: PMC6155955 DOI: 10.1097/md.0000000000012243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Recent findings from an animal experiment suggest a modest association between silymarin and decreased risk of contrast-induced nephropathy. However, the relationship between silymarin and contrast-induced nephropathy in patients with liver cirrhosis remains unclear.From 1997 to 2007, we identified 3019 patients with liver cirrhosis who were administered silymarin and matched them with 3019 patients with liver cirrhosis who were not administered silymarin. Each patient was followed up for a minimum of 4 years. After adjusting for age, gender, hepatitis B, hepatitis C, alcoholic hepatitis, and Charlson comorbidity index, we considered death occurrence and used the Fine and Gray regression models to calculate subdistribution hazard ratios (sHRs) for contrast-induced nephropathy. Sensitivity analyses were also performed using the same model on the subgroups classified by comorbidity.Using the Fine and Gray regression models and with death as the competing risk, we observed that sHR for contrast-induced nephropathy was 0.94-fold higher in the silymarin cohort than in the nonsilymarin cohort (95% confidence interval = 0.61-1.47, P = .791). On the basis of sensitivity analyses results classified by comorbidity, a nonsignificant decrease in risk of contrast-induced nephropathy was found.Silymarin shows no nephron-protective positive effects on contrast-induced nephropathy. Silymarin did not play a nephron-protective role according to Longitudinal Health Insurance Database of Taiwan. Clinical trials are necessary to further assess the nephron-protective effects of silymarin of contrast-induced nephropathy.
Collapse
Affiliation(s)
- Yu-Jui Kuo
- Department of Traditional Chinese Medicine, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation)
- Department of Applied Cosmetology, National Tainan Junior College of Nursing Tainan
| | - Hui-Ping Chang
- Department of Traditional Chinese Medicine, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation)
| | - Yu-Jun Chang
- Epidemiology and Biostatistics Center, Changhua Christian Hospital, Changhua
| | - Hsing-Hsien Wu
- Department of Thoracic Surgery, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation)
| | - Chang-Hua Chen
- Center of Infection Prevention and Control
- Department of Internal Medicine, Changhua Christian Hospital, Changhua
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
115
|
Renner U, Ciato D, Stalla GK. Recent advances in understanding corticotroph pituitary tumor initiation and progression. F1000Res 2018; 7. [PMID: 30228864 PMCID: PMC6117851 DOI: 10.12688/f1000research.14789.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2018] [Indexed: 11/20/2022] Open
Abstract
Cushing’s disease is the most frequent form of hypercortisolism and is caused by hypophyseal corticotroph adenomas secreting excessive amounts of adrenocorticotropic hormone. Most of the tumors develop sporadically and only a limited number of corticotroph adenomas have been found to be associated with different neuroendocrine syndromes or with familial isolated pituitary adenomas. The pathogenic mechanisms of corticotroph adenomas are largely unknown, but the discovered aberrant chaperoning activity of heat shock protein 90 on the one hand and the presence of ubiquitin-specific protease 8 mutations on the other hand partially explained the causes of their development. Corticotroph tumors arise initially as benign microadenomas but with time form invasively growing aggressive macroadenomas which can switch to corticotroph carcinomas in extremely rare cases. The mechanisms through which corticotroph tumors escape from glucocorticoid negative feedback are still poorly understood, as are the processes that trigger the progression of benign corticotroph adenomas toward aggressive and malignant phenotypes. This review summarizes recent findings regarding initiation and progression of corticotroph pituitary tumors.
Collapse
Affiliation(s)
- Ulrich Renner
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Denis Ciato
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Günter K Stalla
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| |
Collapse
|
116
|
Gharbia S, Balta C, Herman H, Rosu M, Váradi J, Bácskay I, Vecsernyés M, Gyöngyösi S, Fenyvesi F, Voicu SN, Stan MS, Cristian RE, Dinischiotu A, Hermenean A. Enhancement of Silymarin Anti-fibrotic Effects by Complexation With Hydroxypropyl (HPBCD) and Randomly Methylated (RAMEB) β-Cyclodextrins in a Mouse Model of Liver Fibrosis. Front Pharmacol 2018; 9:883. [PMID: 30150935 PMCID: PMC6099081 DOI: 10.3389/fphar.2018.00883] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
Silymarin (Sy) shows limited water solubility and poor oral bioavailability. Water-soluble hydroxypropyl (HPBCD) and randomly methylated (RAMEB) β-cyclodextrins were designed to enhance anti-fibrotic efficiency of silymarin in CCl4-induced liver fibrosis in mice. Experimental fibrosis was induced by intraperitoneal injection with 2 ml/kg CCl4 (20% v/v) twice a week, for 7 weeks. Mice were orally treated with 50 mg/kg of Sy-HPBCD, Sy-RAMEB and free silymarin. For assessment of the spontaneous reversion of fibrosis, CCl4 treated animals were investigated after 2 weeks of recovery time. The CCl4 administration increased hepatic oxidative stress, augmented the expression of transforming growth factor-β1 (TGF-β1) and Smad 2/3, and decreased Smad 7 expression. Furthermore, increased α-smooth muscle actin (α-SMA) expression indicated activation of hepatic stellate cells (HSCs), while up-regulation of collagen I (Col I) and matrix metalloproteinases (MMPs) expression led to an altered extracellular matrix enriched in collagen, confirmed as well by trichrome staining and electron microscopy analysis. Treatment with Sy-HPBCD and Sy-RAMEB significantly reduced liver injury, attenuating oxidative stress, restoring antioxidant balance in the hepatic tissue, and significantly decreasing collagen deposits in the liver. The levels of pro-fibrogenic markers' expression were also significantly down-regulated, whereas in the group for spontaneous regression of fibrosis, they remained significantly higher, even at 2 weeks after CCl4 administration was discontinued. The recovery was significantly lower for free silymarin group compared to silymarin/β cyclodextrins co-treatments. Sy-HPBCD was found to be the most potent anti-fibrotic complex. We demonstrated that Sy-HPBCD and Sy-RAMEB complexes decreased extracellular matrix accumulation by inhibiting HSC activation and diminished the oxidative damage. This might occur via the inhibition of TGF-β1/Smad signal transduction and MMP/tissue inhibitor of MMPs (TIMP) rebalance, by blocking the synthesis of Col I and decreasing collagen deposition. These results suggest that complexation of silymarin with HPBCD or RAMEB represent viable options for the its oral delivery, of the flavonoid as a potential therapeutic entity candidate, with applications in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Sami Gharbia
- The Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Cornel Balta
- The Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Hildegard Herman
- The Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Marcel Rosu
- The Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Judit Váradi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Miklós Vecsernyés
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Szilvia Gyöngyösi
- Department of Solid State Physics, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Sorina N Voicu
- Department of Biochemistry and Molecular Biology, The Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Miruna S Stan
- Department of Biochemistry and Molecular Biology, The Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Roxana E Cristian
- Department of Biochemistry and Molecular Biology, The Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, The Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Hermenean
- The Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania.,Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| |
Collapse
|
117
|
Momenkiaei F, Raofie F. Preparation of silybum marianum seeds extract nanoparticles by supercritical solution expansion. J Supercrit Fluids 2018. [DOI: 10.1016/j.supflu.2018.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
118
|
Fischer N, Seo EJ, Efferth T. Prevention from radiation damage by natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 47:192-200. [PMID: 30166104 DOI: 10.1016/j.phymed.2017.11.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/20/2017] [Accepted: 11/12/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Radiotherapy is a mainstay of cancer treatment since decades. Ionizing radiation (IR) is used for destruction of cancer cells and shrinkage of tumors. However, the increase of radioresistance in cancer cells and radiation toxicity to normal tissues are severe concerns. The exposure to radiation generates intracellular reactive oxygen species (ROS), which leads to DNA damage by lipid peroxidation, removal of thiol groups from cellular and membrane proteins, strand breaks and base alterations. HYPOTHESIS Plants have to deal with radiation-induced damage (UV-light of sun, other natural radiation sources). Therefore, it is worth speculating that radioprotective mechanisms have evolved during evolution of life. We hypothesize that natural products from plants may also protect from radiation damage caused as adverse side effects of cancer radiotherapy. METHODS The basis of this systematic review, we searched the relevant literature in the PubMed database. RESULTS Flavonoids, such as genistein, epigallocatechin-3-gallate, epicatechin, apigenin and silibinin mainly act as antioxidant, free radical scavenging and anti-inflammatory compounds, thus, providing cytoprotection in addition to downregulation of several pro-inflammatory cytokines. Comparable effects have been found in phenylpropanoids, especially caffeic acid phenylethylester, curcumin, thymol and zingerone. Besides, resveratrol and quercetin are the most important cytoprotective polyphenols. Their radioprotective effects are mediated by a wide range of mechanisms mainly leading to direct or indirect reduction of cellular stress. Ascorbic acid is broadly used as antioxidant, but it has also shown activity in reducing cellular damage after irradiation mainly due to its antioxidant capabilities. The metal ion chelator, gallic acid, represents another natural product attenuating cellular damage caused by radiation. CONCLUSIONS Some secondary metabolites from plants reveal radioprotective features against cellular damage caused by irradiation. These results warrant further analysis to develop phytochemicals as radioprotectors for clinical use.
Collapse
Affiliation(s)
- Nicolas Fischer
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ean-Jeong Seo
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
119
|
Phyto-phospholipid complexes (phytosomes): A novel strategy to improve the bioavailability of active constituents. Asian J Pharm Sci 2018; 14:265-274. [PMID: 32104457 PMCID: PMC7032241 DOI: 10.1016/j.ajps.2018.05.011] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/14/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Although active constituents extracted from plants show robust in vitro pharmacological effects, low in vivo absorption greatly limits the widespread application of these compounds. A strategy of using phyto-phospholipid complexes represents a promising approach to increase the oral bioavailability of active constituents, which is consist of ‘‘label-friendly” phospholipids and active constituents. Hydrogen bond interactions between active constituents and phospholipids enable phospholipid complexes as an integral part. This review provides an update on four important issues related to phyto-phospholipid complexes: active constituents, phospholipids, solvents, and stoichiometric ratios. We also discuss recent progress in research on the preparation, characterization, structural verification, and increased bioavailability of phyto-phospholipid complexes.
Collapse
|
120
|
Mendez-Sanchez N, Cruz-Ramon VC, Ramirez-Perez OL, Hwang JP, Barranco-Fragoso B, Cordova-Gallardo J. New Aspects of Lipotoxicity in Nonalcoholic Steatohepatitis. Int J Mol Sci 2018; 19:2034. [PMID: 30011790 PMCID: PMC6073816 DOI: 10.3390/ijms19072034] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 02/08/2023] Open
Abstract
NASH is becoming increasingly common worldwide because of the growing global prevalence of obesity and consequently NAFLD. Unfortunately, the mechanism of progression of NAFLD to NASH and then cirrhosis is not completely understood. Several factors, including insulin resistance, inflammation, oxidative stress, lipotoxicity, and bile acid (BA) toxicity, have been reported to be associated with NASH progression. The release of fatty acids from dysfunctional and insulin-resistant adipocytes results in lipotoxicity, which is caused by the ectopic accumulation of triglyceride-derived toxic metabolites and the subsequent activation of inflammatory pathways, cellular dysfunction, and lipoapoptosis. Adipose tissue (AT), especially visceral AT, comprises multiple cell populations that produce adipokines and insulin-like growth factor, plus macrophages and other immune cells that stimulate the development of lipotoxic liver disease. These biomolecules have been recently linked with many digestive diseases and gastrointestinal malignancies such as hepatocellular carcinoma. This made us question what role lipotoxicity has in the natural history of liver fibrosis. Therefore, this review focuses on the close relationship between AT and NASH. A good comprehension of the pathways that are related to dysregulated AT, metabolic dysfunction, and hepatic lipotoxicity will result in the development of prevention strategies and promising therapeutics for patients with NASH.
Collapse
Affiliation(s)
| | | | | | - Jessica P Hwang
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Beatriz Barranco-Fragoso
- Department of Gastroenterology, National Medical Center "20 Noviembre", 03229 Mexico City, Mexico.
| | | |
Collapse
|
121
|
Lee WP, Lan KL, Liao SX, Huang YH, Hou MC, Lan KH. Inhibitory Effects of Amentoflavone and Orobol on Daclatasvir-Induced Resistance-Associated Variants of Hepatitis C Virus. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:835-852. [DOI: 10.1142/s0192415x18500441] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hepatitis C virus (HCV) is recognized as a major causative agent of chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Despite rapid progress in the development of direct-acting antivirals (DAA) against HCV infection in recent years, cost-effective antiviral drugs with more affordable prices still need to be developed. In this study, we screened a library of natural compounds to identify natural HCV inhibitors. The library of the pure compounds extracted from Chinese herbs deposited in the chemical bank of National Research Institute of Chinese Medicine (NRICM), Taiwan was screened in the cell culture-derived HCV (HCVcc) system. We identified the flavone or flavan-based compounds amentoflavone, 7,4[Formula: see text]-dihydroxyflavanone, and orobol with the inhibition of viral entry, replication, and translation of the HCV life cycle. Amentoflavone and orobol also showed inhibitory effects on resistant-associated variants to the NS5A inhibitor daclatasvir. The results of this study have the potential to benefit patients who are intolerant to the adverse effect of pegylated interferon or who harbor resistant strains refractory to treatment by current direct-acting antiviral agents.
Collapse
Affiliation(s)
- Wei-Ping Lee
- Department of Medical Research and Education, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Keng-Li Lan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shi-Xian Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Keng-Hsin Lan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
122
|
Sekkien A, Swilam N, Ebada SS, Esmat A, El-Khatib AH, Linscheid MW, Singab AN. Polyphenols from Tamarix nilotica: LC⁻ESI-MS n Profiling and In Vivo Antifibrotic Activity. Molecules 2018; 23:E1411. [PMID: 29891794 PMCID: PMC6100050 DOI: 10.3390/molecules23061411] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 05/30/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023] Open
Abstract
Tamarix nilotica (Ehrenb.) Bunge (Tamaricaceae), an indigenous plant to the Middle East region, is well-known as a medicinal plant for treating many human ailments. The current study aimed at exploring the polyphenol profile of the alcohol soluble fraction of aqueous T. nilotica extract, assessing its in vivo antifibrotic activity and the possible underlying mechanism, to unravel the impact of quantitative difference of sulphated polyphenols content on the antifibrotic activity of T. nilotca grown in two different habitats. Polyphenol profiling of T. nilotica extracts was performed using HPLC-HRESI-QTOF-MS-MS. The major polyphenol components included sulphated flavonoids, phenolic acids and free aglycones. The antifibrotic activity was evaluated through carbon tetrachloride-induced liver fibrosis in rats. Biochemical evaluations revealed that both fractions ameliorated the increased levels of hepatic aminotransferases, lipid peroxidation, hydroxyproline, α-smooth muscle actin (α-SMA), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2) and nuclear factor kappa B (NF-κB). Moreover, both fractions reduced catalase activity (CAT) and enhanced hepatic glutathione (GSH) content. Histopathological imaging undoubtedly confirmed such results. In conclusion, the T. nilotica polyphenol-rich fraction exhibited potential antifibrotic activity in rats. Significant alterations in GSH levels were recorded based on the sulphated polyphenol metabolite content.
Collapse
Affiliation(s)
- Ahmed Sekkien
- Department of Pharmacognosy, Faculty of Pharmacy, British University in Egypt (BUE), Cairo 11837, Egypt.
| | - Noha Swilam
- Department of Pharmacognosy, Faculty of Pharmacy, British University in Egypt (BUE), Cairo 11837, Egypt.
| | - Sherif S Ebada
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| | - Ahmed Esmat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| | - Ahmed H El-Khatib
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
- Laboratory of Applied Analytical and Environmental Chemistry, Department of Chemistry, Humboldt-Universität zu Berlin, 10099 Berlin, Germany.
| | - Michael W Linscheid
- Laboratory of Applied Analytical and Environmental Chemistry, Department of Chemistry, Humboldt-Universität zu Berlin, 10099 Berlin, Germany.
| | - Abdel Nasser Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
123
|
Martinez-Hurtado J, Calo-Fernandez B, Vazquez-Padin J. Preventing and Mitigating Alcohol Toxicity: A Review on Protective Substances. BEVERAGES 2018; 4:39. [DOI: 10.3390/beverages4020039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
This review covers three fundamental aspects of alcohol consumption and research efforts around the prevention and mitigation of its toxic effects in the human body. First, the sociocultural aspects of alcohol consumption are analysed, including drinking habits and strategies to combat intoxication. Second, we briefly introduce the biochemical aspects of ethanol metabolism and the biochemical pathways leading to its degradation, particularly the activation of toxic response pathways. Finally, we review current evidence and research efforts for finding compounds and substances able to prevent and mitigate the toxic effects of alcohol when over-indulgence has occurred. The toxic effects appear as a time-evolution process based on the stage of intoxication. We explore different compounds and formulations traditionally used to combat alcohol toxicity, as well as state-of-the-art research in the topic for novel molecules and formulations. Although we aimed to categorise which compounds are more effective for a particular level of alcohol intoxication, it is impossible to fully prevent or mitigate toxicity effects by only the compounds in isolation, further research is required to establish the long-term prevention and mitigation from the clinical point of view.
Collapse
|
124
|
Abstract
Acute liver failure (ALF) is a rare but highly fatal condition. The most common causes include drug-induced and viral hepatitis, but other less common etiologies, especially autoimmune hepatitis, Budd-Chiari syndrome, and Wilson disease, need to be considered. Because diagnosis is frequently tied to potential for reversibility of ALF and prognosis, early identification in a timely manner is crucial. Other causes of ALF are more easily recognizable based on specific circumstances, such as ALF in pregnancy or ischemic hepatitis. Ultimately, maintaining a wide differential diagnosis in patients with ALF is essential to identifying the proper treatment and prognosis.
Collapse
Affiliation(s)
- Russell Rosenblatt
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, 1305 York Avenue, 4th Floor, New York, NY 10021, USA
| | - Robert S Brown
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, 1305 York Avenue, 4th Floor, New York, NY 10021, USA.
| |
Collapse
|
125
|
Comparison of the Hepatoprotective Effects of Four Endemic Cirsium Species Extracts from Taiwan on CCl₄-Induced Acute Liver Damage in C57BL/6 Mice. Int J Mol Sci 2018; 19:ijms19051329. [PMID: 29710853 PMCID: PMC5983772 DOI: 10.3390/ijms19051329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 12/19/2022] Open
Abstract
Species of Cirsium (Asteraceae family) have been used in folk hepatoprotective medicine in Taiwan. We collected four Cirsium species—including the aerial part of Cirsium arisanense (CAH), the aerial part of Cirsium kawakamii (CKH), the flower part of Cirsium japonicum DC. var. australe (CJF), and Cirsii Herba (CH)—and then made extractions from them with 70% methanol. We compared the antioxidant contents and activities of these four Cirsium species extracts by a spectrophotometric method and high-performance liquid chromatography⁻photodiode array detector (HPLC-DAD). We further evaluated the hepatoprotective effects of these extracts on CCl₄-induced acute liver damage in C57BL/6 mice. The present study found CAH possesses the highest antioxidant activity among the four Cirsium species, and these antioxidant activities are closely related to phenylpropanoid glycoside (PPG) contents. The extracts decreased serum ALT and AST levels elevated by injection with 0.2% CCl₄. However, only CJF and CH decreased hepatic necrosis. Silibinin decreased serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and hepatic necrosis caused by CCl₄. CJF and CH restored the activities of hepatic antioxidant enzymes and decreased hepatic malondialdehyde (MDA) levels. CJF further restored the expression of hepatic antioxidant enzymes including Cu/Zn-superoxide dismutase (Cu/Zn-SOD), Mn-superoxide dismutase (Mn-SOD), and glutathione S-transferase (GST) proteins. HPLC chromatogram indicated that CKH, CJF, and CH contained silibinin diastereomers (α and β). Only CJF contained diosmetin. Hence, the hepatoprotective mechanism of CJF against CCl₄-induced acute liver damage might be involved in restoring the activities and protein expression of the hepatic antioxidant defense system and inhibiting hepatic inflammation, and these hepatoprotective effects are related to the contents of silibinin diastereomers and diosmetin.
Collapse
|
126
|
Bai ZL, Tay V, Guo SZ, Ren J, Shu MG. Silibinin Induced Human Glioblastoma Cell Apoptosis Concomitant with Autophagy through Simultaneous Inhibition of mTOR and YAP. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6165192. [PMID: 29780826 PMCID: PMC5892302 DOI: 10.1155/2018/6165192] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022]
Abstract
Silibinin, also known as silybin, is the major flavonolignan isolated from Silybum marianum. Although previous reports demonstrated that silibinin exhibits significant tumor suppressor activities in various cancers by promoting cell apoptosis, it was also shown to trigger autophagy to counteract apoptosis induced by exogenous stresses in several types of cells. However, there is no report to address the role of silibinin induced autophagy in human A172 and SR glioblastoma cells. Our study showed that silibinin treatment not only inhibited the metabolic activities of glioblastoma cells but also promoted their apoptosis through the regulation of caspase 3 and PARP-1 in concentration- and time-dependent manners. Meanwhile, silibinin induced autophagy through upregulation of microtubule-associated protein a light chain 3- (LC3-) II. And autophagy inhibition with chloroquine, a lysosomotropic agent, significantly enhanced silibinin induced glioblastoma cell apoptosis. Moreover, silibinin dose-dependently downregulated the phosphorylation levels of mTOR at Ser-2448, p70S6K at Thr-389, and 4E-BP1 at Thr-37/46. Furthermore, the expression of YAP, the downstream effector of Hippo signal pathway, was also suppressed by silibinin. These results suggested that silibinin induced glioblastoma cell apoptosis concomitant with autophagy which might be due to simultaneous inhibition of mTOR and YAP and silibinin induced autophagy exerted a protective role against cell apoptosis in both A172 and SR cells.
Collapse
Affiliation(s)
- Zhuan-Li Bai
- Department of Plastic, Aesthetic and Maxillofacial Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Vincent Tay
- Department of Plastic, Reconstructive and Aesthetic Surgery, Singapore General Hospital, Singapore
| | - Shu-Zhong Guo
- Department of Plastic, Aesthetic and Maxillofacial Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Juan Ren
- Department of Radiotherapy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mao-Guo Shu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
127
|
Diaz JH. Amatoxin-Containing Mushroom Poisonings: Species, Toxidromes, Treatments, and Outcomes. Wilderness Environ Med 2018; 29:111-118. [DOI: 10.1016/j.wem.2017.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 01/03/2023]
|
128
|
García-Ramírez M, Turch M, Simó-Servat O, Hernández C, Simó R. Silymarin prevents diabetes-induced hyperpermeability in human retinal endothelial cells. ACTA ACUST UNITED AC 2018; 65:200-205. [PMID: 29422244 DOI: 10.1016/j.endinu.2017.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Vascular endothelial growth factor (VEGF) plays an essential role in development of diabetic macular edema (DME). While there is evidence suggesting that silymarin, a flavonoid extracted from Silybum marianum, could be useful for prevention and treatment of diabetic nephropathy, no studies have been conducted in diabetic retinopathy (DR). The aim of this study was to assess the effect of silymarin on disruption of inner blood retinal barrier (BRB), the primary cause of DME. MATERIALS AND METHODS Human retinal endothelial cells (HRECs) were cultured under standard (5.5mM D-glucose) and diabetogenic conditions (25mM D-glucose and 25mM D-glucose + recombinant vascular endothelial growth factor [rVEGF, 25mg/mL]). To assess cell viability, three concentrations of silymarin were tested (2, 4 and 10μg/mL). The effect of silymarin on HREC disruption was determined using a dextran (70kD) permeability asssay. RESULTS No differences were found in the viability of HRECs treated with 2 or 4μg/mL of silymarin as compared to untreated cells, but viability significantly decreased after using 10μg/mL. The concentration of 4 μg/mL was therefore selected. Silymarin (4μg/mL) caused a significant decrease in VEGF-induced permeability in both media with 5.5nM (422±58 vs. 600±72 ng/mL/cm2; p<0.03) and 25nM of D-glucose (354 ± 28 vs. 567 ± 102 ng/mL/cm2; p<0.04). DISCUSSION Our results show that silymarin is effective for preventing hyperpermeability induced by diabetic conditions in HRECs. Further studies are needed to assess whether silymarin could be useful to treat DME.
Collapse
Affiliation(s)
- Marta García-Ramírez
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Spain.
| | - Mireia Turch
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Spain
| | - Olga Simó-Servat
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute. Universitat Autònoma de Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Spain.
| |
Collapse
|
129
|
Lutsenko SV, Gromovykh TI, Krasnyuk II, Vasilenko IA, Feldman NB. Antihepatotoxic Activity of Liposomal Silibinin. BIONANOSCIENCE 2018. [DOI: 10.1007/s12668-018-0512-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
130
|
Thakare VN, Patil RR, Oswal RJ, Dhakane VD, Aswar MK, Patel BM. Therapeutic potential of silymarin in chronic unpredictable mild stress induced depressive-like behavior in mice. J Psychopharmacol 2018; 32:223-235. [PMID: 29215318 DOI: 10.1177/0269881117742666] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Silymarin, a plant-derived polyphenolic flavonoid of Silybum marianum, elicited significant antidepressant-like activity in an acute restraint stress model of depression. It improved monoamines, mainly 5-hydroxytryptamine (5-HT) levels in the cortex, dopamine (DA) and norepinephrine (NE) in the cerebellum in mice. The present study was undertaken to explore the antidepressant potential of silymarin in chronic unpredictable mild stress (CUMS) induced depressive-like behavior in mice, and to find out its probable mechanism(s) of action, mainly neurogenesis, neuroinflammation, and/or oxidative stress. The mice were subjected to CUMS for 28 days (4 weeks) and administered with silymarin (100 mg/kg and 200 mg/kg), or fluoxetine or vehicle from days 8 to 28 (3 weeks simultaneously). Animals were evaluated for behavioral changes, such as anhedonia by sucrose preference test, behavioral despair by forced swim test, and exploratory behaviors by an open field test. In addition, neurobiochemical alterations, mainly monoamines, 5-HT, NE, DA, neurotrophic factor BDNF, and cytokines, IL-6, TNF-α, oxidant-antioxidant parameters by determining the malondialdehyde formation (an index of lipid peroxidation process), superoxide dismutase (SOD) and catalase (CAT) activity in hippocampus and cerebral cortex along with serum corticosterone were investigated. Our findings reveal that mice subjected to CUMS exhibited lower sucrose preference, increase immobility time without affecting general locomotion of the animals, and reduce BDNF, 5-HT, NE, and DA level, increased serum corticosterone, IL-6 and TNF-α along with an oxidant-antioxidant imbalance in the hippocampus and cerebral cortex. Silymarin significantly reversed the CUMS-induced changes in the hippocampus and cerebral cortex in mice. Thus, the possible mechanism involved in the antidepressant-like activity of silymarin is correlated to the alleviation of monoaminergic, neurogenesis (enhancing 5-HT, NE, and BDNF levels), and attenuation of inflammatory cytokines system and oxidative stress by modulation of corticosterone response, restoration of antioxidant defense system in cerebral cortex and hippocampus.
Collapse
Affiliation(s)
- Vishnu N Thakare
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India.,4 Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Rajesh R Patil
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India
| | - Rajesh J Oswal
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India
| | | | - Manoj K Aswar
- 3 Department of Pharmacology, Sinhgad Institute of Pharmacy, Nerhe, Pune, India
| | - Bhoomika M Patel
- 4 Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
131
|
Rodriguez JP, Quilantang NG, Lee JS, Lee JM, Kim HY, Shim JS, Lee S. Determination of Silybin B in the Different Parts of Silybum marianum using HPLC-UV. ACTA ACUST UNITED AC 2018. [DOI: 10.20307/nps.2018.24.2.82] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Joyce P. Rodriguez
- Department of Integrative Plant Science, Chung-Ang University, Anseong 17546, Korea
| | - Norman G. Quilantang
- Department of Integrative Plant Science, Chung-Ang University, Anseong 17546, Korea
| | - Ju Sung Lee
- Department of Integrative Plant Science, Chung-Ang University, Anseong 17546, Korea
| | - Jeong Min Lee
- Natural Products Research Team, National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea
| | - Hyun Young Kim
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea
| | - Jae Suk Shim
- Imsil Herbal Medicine Association, Imsil 55955, Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science, Chung-Ang University, Anseong 17546, Korea
| |
Collapse
|
132
|
Sharma D, Sabela MI, Kanchi S, Bisetty K, Skelton AA, Honarparvar B. Green synthesis, characterization and electrochemical sensing of silymarin by ZnO nanoparticles: Experimental and DFT studies. J Electroanal Chem (Lausanne) 2018. [DOI: 10.1016/j.jelechem.2017.11.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
133
|
Wang L, Huang QH, Li YX, Huang YF, Xie JH, Xu LQ, Dou YX, Su ZR, Zeng HF, Chen JN. Protective effects of silymarin on triptolide-induced acute hepatotoxicity in rats. Mol Med Rep 2018; 17:789-800. [PMID: 29115625 PMCID: PMC5780159 DOI: 10.3892/mmr.2017.7958] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 08/31/2017] [Indexed: 02/06/2023] Open
Abstract
Silymarin has been used in the treatment of a number of liver diseases for a long time, but its efficacy in preventing triptolide induced acute hepatotoxicity has not been reported previously. The present study aimed to assess the protective effect of silymarin against triptolide (TP)-induced hepatotoxicity in rats. Rats were orally administrated with silymarin (50, 100 and 200 mg/kg) for 7 days and received intraperitoneal TP (2 mg/kg) on the day 8. Hepatic injuries were comprehensively evaluated in terms of serum parameters, morphological changes, oxidative damage, inflammation and apoptosis. The results demonstrated that TP-induced increases in serum parameters, including alanine transaminase, aspartate aminotransferase, alkaline phosphatase, total cholesterol and γ-glutamyl transpeptidase, which were determined using a biochemical analyzer, and histopathological alterations and hepatocyte apoptosis as determined by hematoxylin and eosin and TUNEL staining, respectively, were prevented by silymarin pretreatment in a dose-dependent manner. TP-induced depletions in the activity of the antioxidant enzymes superoxide dismutase, glutathione peroxidase, glutathione S-transferase and catalase, and glutathione levels, were also significantly reversed by silymarin, as determined using specific kits. Additionally, silymarin dose-dependently exhibited inhibitory effects on malonaldehyde content in the liver. The production of proinflammatory cytokines was investigated using ELISA kits, and the results demonstrated that silymarin dose-dependently inhibited the production of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10 and IL-1β in the liver. To determine the mechanism of silymarin, western blot analysis was performed to investigate the protein expression of phosphorylated (p)-p38 and p-c-Jun N-terminal kinase (JNK) of the TNF-α induced inflammatory response and apoptotic pathways. Silymarin significantly blocked p38 and JNK phosphorylation and activation. Additionally, the expression of the proapoptotic proteins cytochrome c, cleaved caspase-3 and Bcl-2-associated X was also reduced following treatment with silymarin, as determined by ELISA, western blotting and immunohistochemistry, respectively. In conclusion, silymarin was demonstrated to dose-dependently protect rat liver from TP-induced acute hepatotoxicity, with the high dose (200 mg/kg) achieving a superior effect. This protective effect may be associated with the improvement of antioxidant and anti-inflammatory status, as well as the prevention of hepatocyte apoptosis. Therefore, silymarin may have the potential to be applied clinically to prevent TP-induced acute hepatotoxicity.
Collapse
Affiliation(s)
- Lan Wang
- Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Qiong-Hui Huang
- Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yong-Xian Li
- Department of Spine Surgery, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yan-Feng Huang
- Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Jian-Hui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Lie-Qiang Xu
- Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yao-Xing Dou
- Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Zi-Ren Su
- Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, Guangdong 523808, P.R. China
| | - Hui-Fang Zeng
- Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Jian-Nan Chen
- Higher Education Institute and Development Research of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
134
|
St. John TM. Chronic Hepatitis. Integr Med (Encinitas) 2018. [DOI: 10.1016/b978-0-323-35868-2.00021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
135
|
Arif T, Dorjay K, Adil M. Silymarin: An interesting modality in dermatological therapeutics. Indian J Dermatol Venereol Leprol 2018; 84:238-243. [DOI: 10.4103/ijdvl.ijdvl_746_16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
136
|
Darvishi-Khezri H, Salehifar E, Kosaryan M, Karami H, Mahdavi M, Alipour A, Aliasgharian A. Iron-chelating effect of silymarin in patients with β-thalassemia major: A crossover randomised control trial. Phytother Res 2017; 32:496-503. [DOI: 10.1002/ptr.5995] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 10/13/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023]
Affiliation(s)
| | - Ebrahim Salehifar
- Department of Clinical Pharmacology, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| | - Mehrnoush Kosaryan
- Department of Pediatrics, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| | - Hossein Karami
- Department of Pediatrics, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| | - Mohammadreza Mahdavi
- PhD in Medical Genetics, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| | - Abbas Alipour
- Department of Community Medicine, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| | - Aily Aliasgharian
- Student Research Committee, MSc in Medical Microbiology, Thalassemia Research Center, Hemoglobinopathy Institute; Mazandaran University of Medical Sciences; Mazandaran Sari Iran
| |
Collapse
|
137
|
Pendry BA, Kemp V, Hughes MJ, Freeman J, Nuhu HK, Sanchez-Medina A, Corcoran O, Galante E. Silymarin content in Silybum marianum extracts as a biomarker for the quality of commercial tinctures. J Herb Med 2017. [DOI: 10.1016/j.hermed.2017.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
138
|
Yum MJ, Koppula S, Kim JS, Shin GM, Chae YJ, Yoon T, Chun CS, Lee JD, Song M. Protective effects of Ampelopsis brevipedunculata against in vitro hepatic stellate cells system and thioacetamide-induced liver fibrosis rat model. PHARMACEUTICAL BIOLOGY 2017; 55:1577-1585. [PMID: 28395572 PMCID: PMC6130492 DOI: 10.1080/13880209.2017.1311928] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/16/2017] [Accepted: 03/23/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Ampelopsis brevipedunculata Maxim (Vitaceae) is a traditional medicinal herb used for treating liver disorders. OBJECTIVE The hepatoprotective effects of A. brevipedunculata ethanol extract (ABE) was investigated in experimental models of fibrosis. MATERIALS AND METHODS Hepatic stellate cells (HSCs) system in vitro and thioacetamide (TAA)-induced liver fibrosis rat model in vivo were used. Sprague-Dawley rats were divided into five groups of eight each (control, TAA, TAA with ABE 10 mg/kg, ABE 100 mg/kg and silymarin 50 mg/kg groups, respectively). Fibrosis was induced except to the control group by TAA (200 mg/kg, i.p.) twice per week for 13 weeks. ABE and silymarin was administered orally six times per week from the 7th week to the 13th week. RESULTS In HSC-T6 cells, ABE (0.1 mg/mL) and silymarin (0.05 mg/mL) significantly (p < 0.01) induced apoptosis (12.94 ± 5.72% and 14.9 ± 3.8%, respectively) compared with control group (7.51 ± 1.26%). The expression of fibrosis related genes (TGF-β, α-SMA and Col1A1) in HSC-T6 cells were significantly (p < 0.01) downregulated in ABE-treated groups compared with control group. In in vivo studies, ABE (10 and 100 mg/kg) treatment ameliorated the altered levels of serum biomarkers significantly (p < 0.01 and p < 0.001) in TAA-induced groups. Further, ABE (10 and 100 mg/kg) significantly (p < 0.01) attenuated the altered histopathological findings, glutathione content and the accumulation of hydroxyproline. CONCLUSION These results collectively indicate that ABE can potentially be developed as a therapeutic agent in the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Mun Jeong Yum
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
- R&D center Korean Drug Co., Ltd, Seoul, South Korea
| | - Sushruta Koppula
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
- Department of Biotechnology, College of Biomedical and Health Sciences, Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
| | - Jin Seoub Kim
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
| | - Gwang Mo Shin
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
| | - Yun Jin Chae
- Department of Biotechnology, College of Biomedical and Health Sciences, Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
| | - Tony Yoon
- Food One Corp, Deoksan-myeon Jincheon-gun, Chungcheongbuk-do, Korea
| | - Chi Su Chun
- Food One Corp, Deoksan-myeon Jincheon-gun, Chungcheongbuk-do, Korea
| | - Jae Dong Lee
- Department of Internal medicine, School of Medicine, Konkuk University, Chungju, Chungbuk, South Korea
| | - MinDong Song
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
- Department of Biotechnology, College of Biomedical and Health Sciences, Konkuk University, Chungju-si, Chungcheongbuk-do, South Korea
| |
Collapse
|
139
|
Kim JS, Koppula S, Yum MJ, Shin GM, Chae YJ, Hong SM, Lee JD, Song M. Anti-fibrotic effects of Cuscuta chinensis with in vitro hepatic stellate cells and a thioacetamide-induced experimental rat model. PHARMACEUTICAL BIOLOGY 2017; 55. [PMID: 28651481 PMCID: PMC6130704 DOI: 10.1080/13880209.2017.1340965] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
CONTEXT Cuscuta chinensis Lam. (Convolvulaceae) has been used as a traditional herbal remedy for treating liver and kidney disorders. OBJECTIVE Anti-fibrotic effects of C. chinensis extract (CCE) in cellular and experimental animal models were investigated. MATERIALS AND METHODS HSC-T6 cell viability, cell cycle and apoptosis were analysed using MTT assay, flow cytometry and Annexin V-FITC/PI staining techniques. Thioacetamide (TAA)-induced fibrosis model was established using Sprague Dawley rats (n = 10). Control, TAA, CCE 10 (TAA with CCE 10 mg/kg), CCE 100 (TAA with CCE 100 mg/kg) and silymarin (TAA with silymarin 50 mg/kg). Fibrosis was induced by TAA (200 mg/kg, i.p.) twice per week for 13 weeks. CCE and silymarin were administered orally two times per week from the 7th to 13th week. Fibrotic related gene expression (α-SMA, Col1α1 and TGF-β1) was measured by RT-PCR. Serum biomarkers, glutathione (GSH) and hydroxyproline were estimated by spectrophotometer using commercial kits. RESULTS CCE (0.05 and 0.1 mg/mL) and silymarin (0.05 mg/mL) treatment significantly (p < 0.01 and p < 0.001) induced apoptosis (11.56%, 17.52% for CCE; 16.50% for silymarin, respectively) in activated HSC-T6 cells, compared with control group (7.26%). Further, rat primary HSCs showed changes in morphology with CCE 0.1 mg/mL treatment. In in vivo studies, CCE (10 and 100 mg/kg) treatment ameliorated the TAA-induced altered levels of serum biomarkers, fibrotic related gene expression, GSH, hydroxyproline significantly (p < 0.05-0.001) and rescued the histopathological changes. CONCLUSIONS CCE can be developed as a potential agent in the treatment of hepatofibrosis.
Collapse
Affiliation(s)
- Jin Seoub Kim
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
- Department of Infectious Disease, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sushruta Koppula
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
- Department of Biotechnology, College of Biomedical and Health Sciences, Konkuk University, Chungju-si, South Korea
| | - Mun Jeong Yum
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
- R&D Center Korean Drug Co., Ltd, Seoul, South Korea
| | - Gwang Mo Shin
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
| | - Yun Jin Chae
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
| | | | - Jae Dong Lee
- Department of Internal Medicine, School of Medicine, Konkuk University, Chungju-si, South Korea
| | - MinDong Song
- Department of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
- Department of Biotechnology, College of Biomedical and Health Sciences, Konkuk University, Chungju-si, South Korea
- CONTACT MinDong SongDepartment of Applied Life Science, Graduate School of Konkuk University, Chungju-si, South Korea
| |
Collapse
|
140
|
Bijak M. Silybin, a Major Bioactive Component of Milk Thistle (Silybum marianum L. Gaernt.)-Chemistry, Bioavailability, and Metabolism. Molecules 2017; 22:E1942. [PMID: 29125572 PMCID: PMC6150307 DOI: 10.3390/molecules22111942] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 10/28/2017] [Accepted: 11/08/2017] [Indexed: 01/04/2023] Open
Abstract
Milk thistle (Silybum marianum) is a medicinal plant that has been used for thousands of years as a remedy for a variety of ailments. The main component of S. marianum fruit extract (silymarin) is a flavonolignan called silybin, which is not only the major silymarin element but is also the most active ingredient of this extract, which has been confirmed in various studies. This compound belongs to the flavonoid group known as flavonolignans. Silybin's structure consists in two main units. The first is based on a taxifolin, the second a phenyllpropanoid unit, which in this case is conyferil alcohol. These two units are linked together into one structure by an oxeran ring. Since the 1970s, silybin has been regarded in official medicine as a substance with hepatoprotective properties. There is a large body of research that demonstrates silybin's many other healthy properties, but there are still a lack of papers focused on its molecular structure, chemistry, metabolism, and novel form of administration. Therefore, the aim of this paper is a literature review presenting and systematizing our knowledge of the silybin molecule, with particular emphasis on its structure, chemistry, bioavailability, and metabolism.
Collapse
Affiliation(s)
- Michal Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
141
|
Al-Rasheed NM, El-Orabi NF, Fadda LM, Ali HM, Al-Rasheed NM, Bassiouni Y, Aldbass AM. Role of Different Natural Antioxidants in the Modulation of mRNA-expression of Apoptotic Molecules in the Livers of Carbon Tetrachloride-Intoxicated Rats. Anim Biotechnol 2017; 28:253-259. [PMID: 28103144 DOI: 10.1080/10495398.2016.1268621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Overexpression of nuclear factor (NF-κB) or activation of Smad3 by transforming growth factor β (TGF-β1) induced by oncogenes results in overexpression of fibrotic processes and hence cell death. The objective of this study is to examine whether Silymarin (Sil) alone or in combination with Vitamin E (Vit E) and/or Curcumin (Cur) plays a modulatory role against the overexpression of NF-κB, and TGF-β that induced in response to carbon tetrachloride (CCl4) administration. The present work revealed that CCl4 induced elevation of in serum alanine aminotransferase (ALT), Apoptosis regulator (Bax), Smad3, TGF-β, and NF-kB hepatic mRNA expression (using Real-time PCR), administration of Sil alone downregulated these expressions. Treatment with Vit E acid and/ or Cur along with Sil produced best results in this concern. B-cell lymphoma 2 (Bcl-2) expressions were downregulated by CCl4; whereas concurrent treatment of Vit E and/or Cur along with Sil increased its expression. On conclusion, the use of Vit E and/or Cur could potentiate the antiapoptotic action of Sil.
Collapse
Affiliation(s)
- Nouf M Al-Rasheed
- a Pharmacology Department, Faculty of Pharmacy , King Saud University , Riyadh , Kingdom of Saudi Arabia
| | - Naglaa F El-Orabi
- a Pharmacology Department, Faculty of Pharmacy , King Saud University , Riyadh , Kingdom of Saudi Arabia.,b Department of Pharmacology and Toxicology, Faculty of Pharmacy , Suez Canal University , Ismailia , Egypt
| | - Laila Mohamed Fadda
- a Pharmacology Department, Faculty of Pharmacy , King Saud University , Riyadh , Kingdom of Saudi Arabia
| | - Hanaa Mahmoud Ali
- c Department of Genetics and Cytology , National Research Center , Dokki , Egypt.,d Preparatory Year Deanship , King Saud University , Riyadh , Kingdom of Saudi Arabia
| | - Nawal M Al-Rasheed
- a Pharmacology Department, Faculty of Pharmacy , King Saud University , Riyadh , Kingdom of Saudi Arabia
| | - Yieldez Bassiouni
- a Pharmacology Department, Faculty of Pharmacy , King Saud University , Riyadh , Kingdom of Saudi Arabia
| | - Abeer Mohammad Aldbass
- e Biochemistry Department, Faculty of Science , King Saud University , Riyadh , Kingdom of Saudi Arabia
| |
Collapse
|
142
|
Vecchione G, Grasselli E, Cioffi F, Baldini F, Oliveira PJ, Sardão VA, Cortese K, Lanni A, Voci A, Portincasa P, Vergani L. The Nutraceutic Silybin Counteracts Excess Lipid Accumulation and Ongoing Oxidative Stress in an In Vitro Model of Non-Alcoholic Fatty Liver Disease Progression. Front Nutr 2017; 4:42. [PMID: 28971098 PMCID: PMC5609553 DOI: 10.3389/fnut.2017.00042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/30/2017] [Indexed: 01/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidity and mortality. Oxidative stress and release of pro-inflammatory cytokines, such as tumor necrosis factor α (TNFα), are major consequences of hepatic lipid overload, which can contribute to progression of NAFLD to non-alcoholic steatohepatitis (NASH). Also, mitochondria are involved in the NAFLD pathogenesis for their role in hepatic lipid metabolism. Definitive treatments for NAFLD/NASH are lacking so far. Silybin, the extract of the milk thistle seeds, has previously shown beneficial effects in NAFLD. Sequential exposure of hepatocytes to high concentrations of fatty acids (FAs) and TNFα resulted in fat overload and oxidative stress, which mimic in vitro the progression of NAFLD from simple steatosis (SS) to steatohepatitis (SH). The exposure to 50 µM silybin for 24 h reduced fat accumulation in the model of NAFLD progression. The in vitro progression of NAFLD from SS to SH resulted in reduced hepatocyte viability, increased apoptosis and oxidative stress, reduction in lipid droplet size, and up-regulation of IκB kinase β-interacting protein and adipose triglyceride lipase expressions. The direct action of silybin on SS or SH cells and the underlying mechanisms were assessed. Beneficial action of silybin was sustained by changes in expression/activity of peroxisome proliferator-activated receptors and enzymes for FA oxidation. Moreover, silybin counteracted the FA-induced mitochondrial damage by acting on complementary pathways: (i) increased the mitochondrial size and improved the mitochondrial cristae organization; (ii) stimulated mitochondrial FA oxidation; (iii) reduced basal and maximal respiration and ATP production in SH cells; (iv) stimulated ATP production in SS cells; and (v) rescued the FA-induced apoptotic signals and oxidative stress in SH cells. We provide new insights about the direct protective effects of the nutraceutic silybin on hepatocytes mimicking in vitro NAFLD progression.
Collapse
Affiliation(s)
- Giulia Vecchione
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Elena Grasselli
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Federica Cioffi
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Francesca Baldini
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Paulo J. Oliveira
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Vilma A. Sardão
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Katia Cortese
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Antonia Lanni
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Adriana Voci
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Laura Vergani
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| |
Collapse
|
143
|
Marin V, Gazzin S, Gambaro SE, Dal Ben M, Calligaris S, Anese M, Raseni A, Avellini C, Giraudi PJ, Tiribelli C, Rosso N. Effects of Oral Administration of Silymarin in a Juvenile Murine Model of Non-alcoholic Steatohepatitis. Nutrients 2017; 9:1006. [PMID: 28895929 PMCID: PMC5622766 DOI: 10.3390/nu9091006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/07/2017] [Accepted: 09/09/2017] [Indexed: 12/29/2022] Open
Abstract
The increasing prevalence of non-alcoholic fatty liver disease (NAFLD) in adolescents is challenging the global care system. No therapeutic strategies have been defined so far, and changes in the lifestyle remain the only alternative. In this study, we assessed the protective effects of silymarin in a juvenile non-alcoholic steatohepatitis (NASH) model and the in vitro effects on fat-laden human hepatocytes. C57Bl/6 mice were exposed to HFHC diet immediately after weaning. After eight weeks, animals showed histological signs of NASH. Silymarin was added to the HFHC diet, the treatment continued for additional 12 weeks and the effects on BMI, hepatomegaly, visceral fat, lipid profile, transaminases, HOMA-IR, steatosis, inflammation, fibrosis, oxidative stress, and apoptosis were determined. The switch from HFHC to control diet was used to mimic life style changes. In vitro experiments were performed in parallel in human hepatocytes. HFHC diet supplemented with silymarin showed a significant improvement in glycemia, visceral fat, lipid profile, and liver fibrosis. Moreover, it reduced (both in vitro and in vivo) ALT, hepatic inflammation, oxidative stress, and apoptosis. Lifestyle changes restored the control group parameters. The data presented show the beneficial effects of the oral administration of silymarin in the absence of changes in the dietary habits in a juvenile model of NASH.
Collapse
Affiliation(s)
- Veronica Marin
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Silvia Gazzin
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Sabrina E Gambaro
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Matteo Dal Ben
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Sonia Calligaris
- Università di Udine, Dipartimento di Scienze AgroAlimentari, Ambientali e Animali, Via Sondrio 2/A, 33100 Udine, Italy.
| | - Monica Anese
- Università di Udine, Dipartimento di Scienze AgroAlimentari, Ambientali e Animali, Via Sondrio 2/A, 33100 Udine, Italy.
| | - Alan Raseni
- IRCCS Burlo Garofolo Paediatric Hospital, Clinical Chemistry Laboratory, 34100 Trieste, Italy.
| | - Claudio Avellini
- Azienda Ospedaliero-Universitaria "Santa Maria della Misericordia", Dipartimento di Laboratorio, Istituto di Anatomia Patologica, 33100 Udine, Italy.
| | - Pablo J Giraudi
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| | - Natalia Rosso
- Fondazione Italiana Fegato ONLUS-Centro Studi Fegato, Area Science Park Basovizza Bldg, Q SS 14 Km 163,5, Basovizza, 34149 Trieste, Italy.
| |
Collapse
|
144
|
Pashaei-Asl F, Pashaei-Asl R, Khodadadi K, Akbarzadeh A, Ebrahimie E, Pashaiasl M. Enhancement of anticancer activity by silibinin and paclitaxel combination on the ovarian cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1483-1487. [PMID: 28884602 DOI: 10.1080/21691401.2017.1374281] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Ovarian carcinoma is the most lethal cancer among all gynaecological malignancies. One of the most chemotherapy drugs used for ovarian cancer is paclitaxel which induces apoptosis. Paclitaxel has been used for many years. Similar to the most cancers this responds to chemotherapy initially but in a long run, drug resistance happens which fails the treatment procedure. Combination of chemotherapy drugs has been suggested to deal with this issue. Silibinin, a plant extraction, has been used from ancient time in traditional medicine and identified to have powerful antioxidant activity. AIM The aim of this study was to examine the effect of paclitaxel and silibinin combination on SKOV-3 cancer cell line. MATERIALS AND METHODS The human epithelial ovarian cancer cell line, SKOV-3, was cultured and treated with paclitaxel, silibinin and paclitaxel plus silibinin for 48 hours. MTT assay was carried out to determine cell viability. For apoptotic process, we used real-time PCR to study P53 and P21 genes expression after drug treatment and network analysis was performed using Pathway Studio web tool (Elsevier). RESULTS Cell growth was inhibited considerably (p < .05) by combination of paclitaxel and silibinin after 48 hours of treatment. Also silibinin and paclitaxel combination induced apoptosis in SKOV-3 cells. Expression analysis by real-time PCR showed the significant up-regulation of two tumour suppressor genes, P53 and P21 in response to combination of silibinin and paclitaxel. In addition, computational network analysis demonstrated the crosstalk between paclitaxel, silibinin and ovarian cancer. CONCLUSIONS Our results showed that combination of chemotherapy drugs of silibinin and paclitaxel can be more efficient in treatment of ovarian cancer cells.
Collapse
Affiliation(s)
- Fatima Pashaei-Asl
- a Molecular Biology Laboratory, Biotechnology Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Roghiyeh Pashaei-Asl
- b Department of Anatomy , Medical School, Iran University of Medical Science , Tehran , Iran.,c Cellular and Molecular Research Center , Iran University of Medical Sciences , Tehran , Iran
| | - Khodadad Khodadadi
- d Genetic Research Theme, Murdoch Children's Research Institute , Royal Children's Hospital, The University of Melbourne , Melbourne , Australia
| | - Abolfazl Akbarzadeh
- e Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,f Universal Scientific Education and Research Network (USERN) , Tabriz , Iran
| | - Esmaeil Ebrahimie
- g Institute of Biotechnology , Shiraz University , Shiraz , Iran.,h School of Medicine , The University of Adelaide , Adelaide , Australia
| | - Maryam Pashaiasl
- e Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran.,i Women's Reproductive Health Research Centre , Tabriz University of Medical Sciences , Tabriz , Iran.,j Department of Anatomical Sciences, Faculty of Medicine , Tabriz University of Medical Sciences , Iran
| |
Collapse
|
145
|
Esmaeil N, Anaraki SB, Gharagozloo M, Moayedi B. Silymarin impacts on immune system as an immunomodulator: One key for many locks. Int Immunopharmacol 2017; 50:194-201. [PMID: 28672215 DOI: 10.1016/j.intimp.2017.06.030] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022]
Abstract
Silymarin is a flavonoid complex extracted from the Silybum marianum plant. It acts as a strong antioxidant and free radical scavenger by different mechanisms. But in addition to antioxidant effects, silymarin/silybin reveals immunomodulatory affects with both immunostimulatory and immunosuppression activities. Different studies have shown that silymarin has the anti-inflammatory effect through the suppression of NF-κB signaling pathway and TNF-α activation. It also has different immunomodulatory activities in a dose and time-dependent manner. As an immunomodulator agent, silymarin inhibits T-lymphocyte function at low doses while stimulates inflammatory processes at high doses. Studies have shown that silymarin has attenuated autoimmune, allergic, preeclampsia, cancer, and immune-mediated liver diseases and also has suppressed oxidative and nitrosative immunotoxicity. Silymarin also has indicated dual effects on proliferation and apoptosis of different cells. In conclusion, based on the current review, silymarin has a broad spectrum of immunomodulatory functions under different conditions. Recognizing the exact mechanisms of silymarin on cellular and molecular pathways would be very valuable for treatment of immune-mediated diseases. Also further studies are needed to assess the utility of silymarin in protection against autoimmune, cancer, allergic and other diseases in human subjects.
Collapse
Affiliation(s)
- Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Sima Balouchi Anaraki
- Department of Immunology, School of Medicine, Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Gharagozloo
- Department of Pediatrics, Program of Immunology and Allergology, Medical School, Université de Sherbrooke, Canada
| | - Behjat Moayedi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
146
|
Elyasi S, Shojaee FSR, Allahyari A, Karimi G. Topical Silymarin Administration for Prevention of Capecitabine-Induced Hand-Foot Syndrome: A Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. Phytother Res 2017; 31:1323-1329. [PMID: 28635153 DOI: 10.1002/ptr.5857] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
Hand-foot syndrome (HFS) is a frequent dose-limiting adverse reaction of capecitabine in patient with gastrointestinal cancers. Silymarin is a polyphenolic flavonoid extracted from the Silybum marianum that exhibits strong antioxidant and antiinflammatory activities. In this study, we evaluated silymarin efficacy in prevention of capecitabine-induced HFS in patients with gastrointestinal cancers, as the first human study. During this pilot, randomized, double-blinded, placebo-controlled clinical trial, the effect of silymarin gel 1%, which is applied on the palms and soles twice daily starting at the first day of chemotherapy for 9 weeks, on HFS occurrence was assessed. Forty patients fulfilled the inclusion criteria assigned to the silymarin or placebo group. World Health Organization HFS grading scale scores were recorded at baseline and every 3 weeks during these 9 weeks. The median WHO HFS scores were significantly lower in silymarin group at the end of the 9th week (p < 0.05). The scores increased significantly in both placebo and silymarin groups during chemotherapy, but there was a delay for HFS development and progression in silymarin group. Prophylactic administration of silymarin topical formulation could significantly reduce the severity of capecitabine-induced HFS and delays its occurrence in patients with gastrointestinal cancer after 9 weeks of application. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sepideh Elyasi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Abolghasem Allahyari
- Hematology-Oncology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
147
|
Bayat A, Fattahi A. The free radical scavenging activity of lespedezacoumestan toward ˙OH radical: A quantum chemical and computational kinetics study. J PHYS ORG CHEM 2017. [DOI: 10.1002/poc.3755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ahmad Bayat
- Department of Chemistry; Sharif University of Technology; Tehran Iran
| | - Alireza Fattahi
- Department of Chemistry; Sharif University of Technology; Tehran Iran
| |
Collapse
|
148
|
Zheng W, Feng Z, Lou Y, Chen C, Zhang C, Tao Z, Li H, Cheng L, Ying X. Silibinin protects against osteoarthritis through inhibiting the inflammatory response and cartilage matrix degradation in vitro and in vivo. Oncotarget 2017; 8:99649-99665. [PMID: 29245931 PMCID: PMC5725122 DOI: 10.18632/oncotarget.20587] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. Silibinin, a polyphenolic flavonoid derived from fruits and seeds of Silybum marianum, has been reported to possess various potent beneficial biological effects, such as antioxidant, anti-cancer, hepatoprotective and anti-inflammatory activities. However, the anti-inflammatory effects of silibinin on OA have not been reported. This study aimed to assess the effects of silibinin on OA both in vitro and in vivo. In this study, we found that silibinin significantly inhibited the nterleukin-1β (IL-1β)-induced production of nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α) and IL-6, expression of cyclooxygenase2 (COX-2), inducible nitric oxide synthase (iNOS), matrix metalloproteinase-1 (MMP-1), MMP-3, MMP-13, a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4) and ADAMTS-5, degradation of aggrecan and collagen-II in human OA chondrocytes. Furthermore, silibinin dramatically suppressed IL-1β-stimulated phosphatidylinositol 3 kinase/ protein kinase B (PI3K/Akt) phosphorylation and nuclear factor-kappa B (NF-kB) activation in human OA chondrocytes. In addition, treatment of silibinin not only prevented the destruction of cartilage and the thickening of subchondral bone but also relieved synovitis in mice OA models. Also, the immunohistochemistry results showed that silibinin significantly decreased the expression of MMP-13 and ADAMTS-5 and increased the expression of collagen-II and aggrecan in mice OA. Taken together, these results suggest that silibinin may be a potential agent in the treatment of OA.
Collapse
Affiliation(s)
- Wenhao Zheng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhenhua Feng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yiting Lou
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chunhui Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chuanxu Zhang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhenyu Tao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hang Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liang Cheng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaozhou Ying
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
149
|
Asgarshirazi M, Shariat M, Sheikh M. Comparison of efficacy of folic acid and silymarin in the management of antiepileptic drug induced liver injury: a randomized clinical trial. Hepatobiliary Pancreat Dis Int 2017; 16:296-302. [PMID: 28603098 DOI: 10.1016/s1499-3872(16)60142-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Liver injury associated with antiepileptic drugs accounts for a large proportion of drug-induced liver injuries (DILI) in children. Although withdrawal of the causative agent is the only proved treatment for DILI, in some clinical situations it is not possible. Recent studies have reported promising results of using hepatoprotective drugs with antioxidant actions for the management of DILI. This study aimed to evaluate the efficacy of folic acid versus silymarin treatment in relation to decreasing liver enzymes in patients with DILI due to antiepileptic therapy. METHODS This randomized, open-label, clinical trial evaluated 55 children with epilepsy who were on antiepileptic treatment and experienced DILI. The children were randomized to receive either silymarin (5 mg/kg per day) or folic acid (1 mg per day) for one month and were followed up for three months. RESULTS Liver enzymes significantly decreased in both groups. The decrease trend in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were stronger in the folic acid group compared to silymarin group (P=0.04 and P=0.007, respectively). At the end of the study patients in the folic acid group had significantly lower ALT (P=0.04), AST (P=0.02), and gamma-glutamyl transferase (GGT) (P<0.001) levels and also higher percentage of normal ALT (30.7% vs 3.4%, P=0.009) and AST (42.3% vs 0%, P<0.001), and GGT (23.1% vs 0%, P=0.008) values compared to the patients in the silymarin group. No rebound elevations in ALT, AST and GGT levels or adverse reactions were noted in neither of the study groups. CONCLUSION Although both treatments were safe and effective in decreasing liver enzymes, folic acid seems to be superior to silymarin in the management of DILI.
Collapse
Affiliation(s)
| | | | - Mahdi Sheikh
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
150
|
Onaolapo AY, Abdusalam SZ, Onaolapo OJ. Silymarin attenuates aspartame-induced variation in mouse behaviour, cerebrocortical morphology and oxidative stress markers. PATHOPHYSIOLOGY 2017; 24:51-62. [DOI: 10.1016/j.pathophys.2017.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/30/2016] [Accepted: 01/16/2017] [Indexed: 12/11/2022] Open
|