1
|
Han YM, Ahn HR, Lee DY, Song MY, Lee SW, Jang YK, Jeon BY, Kim EH. Therapeutic Potential of Hongjam in A Diethylnitrosamine and Thioacetamide-induced Hepatocellular Carcinoma Mouse Model. J Cancer Prev 2024; 29:165-174. [PMID: 39790225 PMCID: PMC11706731 DOI: 10.15430/jcp.24.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common and lethal type of primary liver cancer, frequently arising from chronic liver injury and inflammation. Despite treatment advancements, HCC prognosis remains poor, emphasizing the need for effective preventive and therapeutic strategies. This study investigates the hepatoprotective and anti-tumor effects of Hongjam, a steamed freeze-dried silkworm powder, in a diethylnitrosamine (DEN) and thioacetamide (TAA)-induced HCC mouse model. Mice were administered DEN intraperitoneally for 8 weeks, followed by TAA in drinking water for 9 weeks, with Hongjam supplementation (0.01, 0.1, and 1 g/kg) provided daily through food. Hongjam markedly reduced the tumor incidence, the size, and the histological lesions compared to the DEN/TAA group. Serum biochemical analysis revealed reduction in liver damage markers, including alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, and total bilirubin, with a notable decrease in total bilirubin surpassing. Immunohistochemical and Western blot analyses demonstrated that Hongjam downregulated expression of proliferation markers, including Ki67, phosphorylation of protein kinase B, and proliferating cell nuclear antigen, while upregulating the pro-apoptotic protein Bcl-2-associated X protein, indicating its dual role in suppressing proliferation and promoting apoptosis. Furthermore, Hongjam inhibited angiogenesis by suppressing the expression of key markers, including interleukin 6, VEGF, hypoxia-inducible factor-1 subunit alpha, platelet-derived growth factor subunit beta, matrix metalloproteinase-2, and cluster of differentiation 31, thereby disrupting the tumor microenvironment. These findings suggest that Hongjam exerts multifaceted protective effects against HCC by targeting proliferation, apoptosis, and angiogenesis pathways, while also mitigating liver damage. This study highlights the potential of Hongjam as a functional food or a complementary therapeutic agent for HCC prevention and management.
Collapse
Affiliation(s)
- Young-Min Han
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Hye-Rin Ahn
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Da-Young Lee
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Moon-Young Song
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Seung-Won Lee
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | | | | | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| |
Collapse
|
2
|
El-Gendy HF, Khalifa HK, Omran A, Korany RMS, Selim S, Hussein E, Alhotan RA, Ayyoub A, Masoud SR. Unveiling the Potential of Silymarin, Spirulina platensis, and Chlorella vulgaris towards Cardiotoxicity via Modulating Antioxidant Activity, Inflammation, and Apoptosis in Rats. Life (Basel) 2024; 14:1289. [PMID: 39459589 PMCID: PMC11509764 DOI: 10.3390/life14101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This study assessed the possible pharmacological effects of Chlorella vulgaris (Cg), Spirulina platensis (St), and silymarin (Sl) against thioacetamide (TA)-induced cardiotoxicity in rats, with a focus on their antioxidant, cardioprotective, and anti-inflammatory properties. The following is the random grouping of sixty male rats into six groups of ten animals each: the control (negative control), TA-intoxicated group (positive control; 300 mg/kg body weight (BW)), Sl + TA group (100 mg Sl/kg BW + TA), St + TA group (400 mg St/kg BW + TA), Cg + TA (400 mg Cg/kg BW + TA), and St + Cg + TA group (400 St + 400 Cg mg/kg BW + TA) were all administered for 30 days. At the start of the study, groups 2 through 6 were administered TA intraperitoneally at a dosage of 300 mg/kg BW for two consecutive days, with a 24 h gap between each dose, to induce cardiac damage. Blood samples were obtained to measure hematological parameters and perform biochemical assays, including lipid profiles and cardiac enzymes. For histopathology and immunohistochemistry determination, tissue samples were acquired. The current findings showed that TA injection caused hematological alterations and cardiac injury, as evidenced by greater serum levels of troponin I, creatine kinase-MB, and total creatine kinase (p < 0.05), as well as significantly elevated serum malondialdehyde and decreased serum total antioxidant capacity (p < 0.05) concentrations. Moreover, an increase in blood low-density lipoprotein and total cholesterol concentration (p < 0.05) was recorded in the TA group. There were alterations in the heart tissue's histological structure of the TA group compared to the control ones. These alterations were characterized by vacuolar degeneration of myocytes, loss of cross striation, coagulative necrosis, and fibrosis of interstitial tissue, which was ameliorated by the supplementation of SI, St, and Cg. The TA-intoxicated group showed weak expression of B-cell lymphoma protein 2 (p < 0.05) and strong immunoreactivity of tumor necrosis factor-α and B-cell lymphoma protein 2-associated X (p < 0.05). However, the groups receiving Sl, St, and Cg experienced the opposite. The administration of Sl, St, Cg, and St + Cg along with TA significantly improved and restored (p < 0.05) erythrogram indices, including RBCs, hemoglobin, total leukocytic count, lymphocytes, and monocyte, to the normal control values. The administration of Sl, St, and Cg alleviated the cardiotoxicity caused by TA via reducing oxidative stress, inflammatory markers, and apoptosis in heart tissue. In summary, the current findings suggest that the treatment with Sl, St, and Cg was beneficial in ameliorating and reducing the cardiotoxicity induced by TA in rats.
Collapse
Affiliation(s)
- Hanem F. El-Gendy
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt;
| | - Hanem K. Khalifa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt;
| | - Ahmed Omran
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt;
| | - Reda M. S. Korany
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo 12211, Egypt;
- Department of Pathology, Faculty of Veterinary Medicine, Egyptian Chinese University, Cairo 11765, Egypt
| | - Shaimaa Selim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Menoufia University, Shibin El-Kom 32514, Egypt
| | - Eman Hussein
- Department of Poultry and Fish Production, Faculty of Agriculture, University of Menoufia, Shibin El-Kom 32514, Egypt;
| | - Rashed A. Alhotan
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Anam Ayyoub
- College of Life Sciences, Northwest A & F University, Yangling District, Xianyang 712100, China;
| | - Shimaa R. Masoud
- Department of Physiology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt;
| |
Collapse
|
3
|
Irani Mokhtari G, Azaran A, Rajahi E, Hesam S, Dehghani Ghahfarokhi A, Makvandi M. Prevalence of JC polyomavirus among rheumatoid arthritis and systemic lupus erythematosus patients and its correlation with vitamin D levels. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:676-683. [PMID: 39534291 PMCID: PMC11551653 DOI: 10.18502/ijm.v16i5.16803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Background and Objectives Vitamin D deficiency in viral infection associated with autoimmune diseases is well documented. This study assessed the prevalence of JC virus in patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), and its correlation with vitamin D level. Materials and Methods Serum and urine samples were collected from 50 patients with RA and SLE. DNA was extracted and subjected to PCR test. Positive PCR products were sequenced, phylogenetic tree was constructed to determine the JC virus genotype. The patient's vitamin D level was evaluated. Results Of 50 patients, 19 (38%) were diagnosed as RA, and 31 (62%) were identified as SLE. JC virus DNA was detected in 17 (34%) patients' urine samples including 5 (26.3%) RA and 12 (38.7%) SLE cases. JC virus DNA was detected 2 (4%) in patients' serum samples (one RA. and one SLE). JC virus genotype 3A was dominant. Interestingly, the SLE patients with positive JC virus showed lowered vitamin D compared to patients with negative JC virus (P<0.005). Conclusion Given the high rate of JC virus, DNA detection and susceptibility of patients for PML development, it is recommended that detection of JC virus DNA and vitamin D level should be implemented for patients with RA/SLE prior to treatment.
Collapse
Affiliation(s)
- Golnaz Irani Mokhtari
- Cellular and Molecular Research Center, Ahvaz Jundishapur University, Ahvaz, Iran
- Department of Virology, Ahvaz Jundishapur University, Ahvaz, Iran
| | - Azarakhsh Azaran
- Department of Virology, Ahvaz Jundishapur University, Ahvaz, Iran
| | - Elham Rajahi
- Department of Rheumatology, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeed Hesam
- Department of Biostatistics and Epidemiology, Faculty of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Manoochehr Makvandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University, Ahvaz, Iran
- Department of Virology, Ahvaz Jundishapur University, Ahvaz, Iran
| |
Collapse
|
4
|
Lv YF, Xie CS, Liu ZX, Kang MD, Liu Y, Liao ZQ, Ji YL, Zhao R, Li YS, Wei XY, Luo RG, Tang Q. Sevelamer reverses liver fibrosis by deactivation of hepatic stellate cells. Biochem Pharmacol 2024; 222:116121. [PMID: 38461906 DOI: 10.1016/j.bcp.2024.116121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/22/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Liver fibrosis is a chronic liver disease characterized by a progressive wound healing response caused by chronic liver injury. Currently, there are no approved clinical treatments for liver fibrosis. Sevelamer is used clinically to treat hyperphosphatemia and has shown potential therapeutic effects on liver diseases. However, there have been few studies evaluating the therapeutic effects of sevelamer on liver fibrosis, and the specific mechanisms are still unclear. In this study, we investigated the antifibrotic effects of sevelamer-induced low inorganic phosphate (Pi) stress in vitro and in vivo and analyzed the detailed mechanisms. We found that low Pi stress could inhibit the proliferation of activated hepatic stellate cells (HSCs) by promoting apoptosis, effectively suppressing the migration and epithelial-mesenchymal transition (EMT) of hepatic stellate cells. Additionally, low Pi stress significantly increased the antioxidant stress response. It is worth noting that low Pi stress indirectly inhibited the activation and migration of HSCs by suppressing transforming growth factor β (TGF-β) expression in macrophages. In a rat model of liver fibrosis, oral administration of sevelamer significantly decreased blood phosphorus levels, improved liver function, reduced liver inflammation, and increased the antioxidant stress response in the liver. Our study revealed that the key mechanism by which sevelamer inhibited liver fibrosis involved binding to gastrointestinal phosphate, resulting in a decrease in blood phosphorus levels, the downregulation of TGF-β expression in macrophages, and the inhibition of HSC migration and fibrosis-related protein expression. Therefore, our results suggest that sevelamer-induced low Pi stress can attenuate hepatic stellate cell activation and inhibit the progression of liver fibrosis, making it a potential option for the treatment of liver fibrosis and other refractory chronic liver diseases.
Collapse
Affiliation(s)
- Yang-Feng Lv
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China; Institute for Advanced Study, Nanchang University, Nanchang 330031, China
| | - Chuan-Sheng Xie
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China
| | - Zhi-Xing Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Mei-Diao Kang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China
| | - Yue Liu
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China
| | - Zi-Qiang Liao
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China; Institute for Advanced Study, Nanchang University, Nanchang 330031, China
| | - Yu-Long Ji
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China
| | - Rui Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yan-Shu Li
- Jiangxi Center of Medical Device Testing, Nanchang 330029, China
| | - Xiao-Yong Wei
- Department of Hepatobiliary Surgery, Jiangxi Provincial Cancer Hospital, Nanchang 330029, China
| | - Rong-Guang Luo
- Department of Medical Imaging and Interventional Radiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qun Tang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang 330031, China; Institute for Advanced Study, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
5
|
Mulè S, Ferrari S, Rosso G, Brovero A, Botta M, Congiusta A, Galla R, Molinari C, Uberti F. The Combined Antioxidant Effects of N-Acetylcysteine, Vitamin D3, and Glutathione from the Intestinal-Neuronal In Vitro Model. Foods 2024; 13:774. [PMID: 38472887 DOI: 10.3390/foods13050774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic oxidative stress has been consistently linked to age-related diseases, conditions, and degenerative syndromes. Specifically, the brain is the organ that significantly contributes to declining quality of life in ageing. Since the body cannot completely counteract the detrimental effects of oxidative stress, nutraceuticals' antioxidant properties have received significant attention in recent years. This study assesses the potential health benefits of a novel combination of glutathione, vitamin D3, and N-acetylcysteine. To examine the combination's absorption and biodistribution and confirm that it has no harmful effects, the bioavailability of the mixture was first evaluated in a 3D model that mimicked the intestinal barrier. Further analyses on the blood-brain barrier was conducted to determine the antioxidant effects of the combination in the nervous system. The results show that the combination reaches the target and successfully crosses the blood-brain and intestinal barriers, demonstrating enhanced advantages on the neurological system, such as a reduction (about 10.5%) in inflammation and enhancement in cell myelination (about 20.4%) and brain tropism (about 18.1%) compared to the control. The results support the cooperative effect of N-acetylcysteine, vitamin D3, and glutathione to achieve multiple health benefits, outlining the possibility of an alternative nutraceutical approach.
Collapse
Affiliation(s)
- Simone Mulè
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Sara Ferrari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Giorgia Rosso
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Arianna Brovero
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy
| | - Mattia Botta
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Alessia Congiusta
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Rebecca Galla
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
- Noivita S.r.l.s., Spin Off of University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Claudio Molinari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| | - Francesca Uberti
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, University of Piemonte Orientale, UPO, 13100 Vercelli, Italy
| |
Collapse
|
6
|
Jiang R, Zhou Y, Han L, Hong Z. Serum vitamin D is associated with ultrasound-defined hepatic fibrosis. Clin Res Hepatol Gastroenterol 2023; 47:102228. [PMID: 37865224 DOI: 10.1016/j.clinre.2023.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Evidences from population-based investigations on the exact relationship between vitamin D and the severity of liver fibrosis remain debated and conflicting. Here, we aim to explore the relationship between serum vitamin D and ultrasound-defined advanced hepatic fibrosis in the US participants with nonalcoholic fatty liver disease (NAFLD). METHODS In the retrospective study, individuals with intact information on interesting variables from the 2017-2018 National Health and Nutrition Examination Survey (NHANES) were included. NAFLD was diagnosed on the basis of controlling attenuation parameter (CAP) value≥ 274 dB/m without causes of other chronic hepatic diseases. We identified advanced fibrosis grades (F2) by liver stiffness measurement (LSM) score of ≥ 8.2 kPa in NAFLD patients. The impact of elevated serum vitamin D on the prevalence of hepatic fibrosis was assessed by multivariate logistic regression models on the basis of the NHANES recommended weights. RESULTS The study involved 1624 subjects with NAFLD in total, and 305 (18.28 %, weighted%) of whom were diagnosed with advanced hepatic fibrosis according to the definition based on parameters obtained from vibration controlled transient elastography (VCTE). In the multivariate logistic regression analysis, serum vitamin D presented a negative relationship to hepatic fibrosis with lower odds in patients with hepatic steatosis after being adjusted for potential confounding factors (fully adjusted: OR=0.47, 95 % CI: 0.24-0.90, p = 0.034). Our subgroup analysis revealed that the inverse relationship was still existed in males (fully adjusted: OR = 0.34, 95 % CI: 0.17-0.70, p = 0.014), non-obese subjects (fully adjusted: OR = 0.20, 95 % CI: 0.04-0.89, p = 0.042) and participants below 60 years (fully adjusted: OR = 0.43, 95 % CI: 0.21-0.90, p = 0.033), whereas in models adjusted for the potential confounding factors, no statistically significant correlation was noted in females, obese subjects or subjects with age≥ 60 years. CONCLUSIONS This large population-based investigation indicated that elevated serum vitamin D reduced the onset of advanced fibrosis diagnosed by ultrasound in males, non-obese subjects and younger participants with NAFLD.
Collapse
Affiliation(s)
- Rong Jiang
- Jiangsu Health Vocational College, Nanjing, Jiangsu 210000, China.
| | - Yichao Zhou
- Department of Occupation Disease Prevention and Cure, Changzhou Wujin District Center for Disease Control and Prevention, Changzhou, Jiangsu 213100, China
| | - Lei Han
- Department of Occupation Disease Prevention and Cure, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu 210000, China.
| | - Zhen Hong
- Jiangsu Health Vocational College, Nanjing, Jiangsu 210000, China.
| |
Collapse
|
7
|
Turk A, Kuloglu T, Karadag A, Ozcan Metin T. Effects of Vitamin D on Asprosin Immunoreactivity Against Cyclophosphamide-Induced Liver Injury in Rats. Cureus 2023; 15:e46711. [PMID: 37822688 PMCID: PMC10562880 DOI: 10.7759/cureus.46711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 10/13/2023] Open
Abstract
Background Cyclophosphamide (CP), commonly used as an anticarcinogenic drug, has the potential to induce detrimental effects on multiple tissues, including the liver. Asprosin, which is a glucogenic adipokine, induces the liver to secrete glucose, thus contributing to the maintenance of homeostasis. This study aims to investigate the immunoreactivity of asprosin in the liver tissue of rats exposed to CP administration, as well as the changes in its levels due to the supplementation of Vitamin D (Vit D). Materials and methods Four experimental groups were formed, including control, Vit D (200 IU/kg), CP (200 mg/kg), and Vit D+ CP. Histopathological analysis was carried out by employing staining methods on liver tissues. These techniques encompassed the application of hematoxylin-eosin (H&E), Masson's trichrome, and periodic acid Schiff (PAS). Through the application of spectrophotometric methods, concentrations of malondialdehyde (MDA), total antioxidant status (TAS), total oxidant status (TOS), and asprosin were determined. Furthermore, apoptotic cells were identified by the terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) method, and the asprosin immunoreactivity was determined by immunohistochemistry. Results Under light microscope examination, the histopathological damage was found to be more notable in the CP group compared to the control group. Moreover, a decrease was observed in serum and tissue asprosin levels, while an increase was noted in the count of apoptotic cells, along with elevated MDA and TOS levels. However, in the CP+Vit D group, Vit D administration alleviated histopathological damage. Notably, there were significant increases in TAS and asprosin levels, accompanied by reductions in both MDA and TOS levels. Conclusions The effect of CP on liver tissue was observed to result in damage and a reduction in asprosin levels. Vit D supplementation revealed elevated asprosin levels and a distinct protective effect on the tissue. Considering the association between asprosin and liver injury induced by CP, further research is needed to elucidate the mechanisms that underlie the effect of asprosin on tissues. When combined with Vit D, asprosin holds promise for potential clinical applications as a therapeutic target.
Collapse
Affiliation(s)
- Ahmet Turk
- Department of Histology and Embryology, Faculty of Medicine, Adiyaman University, Adiyaman, TUR
| | - Tuncay Kuloglu
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazig, TUR
| | - Abdullah Karadag
- Department of Physiology, Faculty of Medicine, Adiyaman University, Adiyaman, TUR
| | - Tuba Ozcan Metin
- Department of Histology and Embryology, Faculty of Medicine, Kahramanmaras Sutcu Imam University, Kahramanmaras, TUR
| |
Collapse
|
8
|
Zhang CY, Liu S, Yang M. Treatment of liver fibrosis: Past, current, and future. World J Hepatol 2023; 15:755-774. [PMID: 37397931 PMCID: PMC10308286 DOI: 10.4254/wjh.v15.i6.755] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 06/25/2023] Open
Abstract
Liver fibrosis accompanies the progression of chronic liver diseases independent of etiologies, such as hepatitis viral infection, alcohol consumption, and metabolic-associated fatty liver disease. It is commonly associated with liver injury, inflammation, and cell death. Liver fibrosis is characterized by abnormal accumulation of extracellular matrix components that are expressed by liver myofibroblasts such as collagens and alpha-smooth actin proteins. Activated hepatic stellate cells contribute to the major population of myofibroblasts. Many treatments for liver fibrosis have been investigated in clinical trials, including dietary supplementation (e.g., vitamin C), biological treatment (e.g., simtuzumab), drug (e.g., pegbelfermin and natural herbs), genetic regulation (e.g., non-coding RNAs), and transplantation of stem cells (e.g., hematopoietic stem cells). However, none of these treatments has been approved by Food and Drug Administration. The treatment efficacy can be evaluated by histological staining methods, imaging methods, and serum biomarkers, as well as fibrosis scoring systems, such as fibrosis-4 index, aspartate aminotransferase to platelet ratio, and non-alcoholic fatty liver disease fibrosis score. Furthermore, the reverse of liver fibrosis is slowly and frequently impossible for advanced fibrosis or cirrhosis. To avoid the life-threatening stage of liver fibrosis, anti-fibrotic treatments, especially for combined behavior prevention, biological treatment, drugs or herb medicines, and dietary regulation are needed. This review summarizes the past studies and current and future treatments for liver fibrosis.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Shuai Liu
- Department of Radiology,The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
9
|
Shi L, Zhou L, Han M, Zhang Y, Zhang Y, Yuan XX, Lu HP, Wang Y, Yang XL, Liu C, Wang J, Liang P, Liu SA, Liu XJ, Cheng J, Lin SM. Calcitriol attenuates liver fibrosis through hepatitis C virus nonstructural protein 3-transactivated protein 1-mediated TGF β1/Smad3 and NF-κB signaling pathways. World J Gastroenterol 2023; 29:2798-2817. [PMID: 37274069 PMCID: PMC10237113 DOI: 10.3748/wjg.v29.i18.2798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/08/2023] [Accepted: 04/10/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Hepatic fibrosis is a serious condition, and the development of hepatic fibrosis can lead to a series of complications. However, the pathogenesis of hepatic fibrosis remains unclear, and effective therapy options are still lacking. Our group identified hepatitis C virus nonstructural protein 3-transactivated protein 1 (NS3TP1) by suppressive subtractive hybridization and bioinformatics analysis, but its role in diseases including hepatic fibrosis remains undefined. Therefore, additional studies on the function of NS3TP1 in hepatic fibrosis are urgently needed to provide new targets for treatment.
AIM To elucidate the mechanism of NS3TP1 in hepatic fibrosis and the regulatory effects of calcitriol on NS3TP1.
METHODS Twenty-four male C57BL/6 mice were randomized and separated into three groups, comprising the normal, fibrosis, and calcitriol treatment groups, and liver fibrosis was modeled by carbon tetrachloride (CCl4). To evaluate the level of hepatic fibrosis in every group, serological and pathological examinations of the liver were conducted. TGF-β1 was administered to boost the in vitro cultivation of LX-2 cells. NS3TP1, α-smooth muscle actin (α-SMA), collagen I, and collagen III in every group were examined using a Western blot and real-time quantitative polymerase chain reaction. The activity of the transforming growth factor beta 1 (TGFβ1)/Smad3 and NF-κB signaling pathways in each group of cells transfected with pcDNA-NS3TP1 or siRNA-NS3TP1 was detected. The statistical analysis of the data was performed using the Student’s t test.
RESULTS NS3TP1 promoted the activation, proliferation, and differentiation of hepatic stellate cells (HSCs) and enhanced hepatic fibrosis via the TGFβ1/Smad3 and NF-κB signaling pathways, as evidenced by the presence of α-SMA, collagen I, collagen III, p-smad3, and p-p65 in LX-2 cells, which were upregulated after NS3TP1 overexpression and downregulated after NS3TP1 interference. The proliferation of HSCs was lowered after NS3TP1 interference and elevated after NS3TP1 overexpression, as shown by the luciferase assay. NS3TP1 inhibited the apoptosis of HSCs. Moreover, both Smad3 and p65 could bind to NS3TP1, and p65 increased the promoter activity of NS3TP1, while NS3TP1 increased the promoter activity of TGFβ1 receptor I, as indicated by coimmunoprecipitation and luciferase assay results. Both in vivo and in vitro, treatment with calcitriol dramatically reduced the expression of NS3TP1. Calcitriol therapy-controlled HSCs activation, proliferation, and differentiation and substantially suppressed CCl4-induced hepatic fibrosis in mice. Furthermore, calcitriol modulated the activities of the above signaling pathways via downregulation of NS3TP1.
CONCLUSION Our results suggest that calcitriol may be employed as an adjuvant therapy for hepatic fibrosis and that NS3TP1 is a unique, prospective therapeutic target in hepatic fibrosis.
Collapse
Affiliation(s)
- Liu Shi
- Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Li Zhou
- China-Japan Friendship Hospital, Department of Infectious Disease China-Japan Friendship Hospital, Beijing 100029, China
| | - Ming Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yu Zhang
- The Division of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yang Zhang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiao-Xue Yuan
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Hong-Ping Lu
- Institute of Liver Diseases, Beijing Pan-Asia Tongze Institute of Biomedicine Co., Ltd, Beijing 100015, China
| | - Yun Wang
- The Division of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xue-Liang Yang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Chen Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jun Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Pu Liang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Shun-Ai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiao-Jing Liu
- Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Shu-Mei Lin
- Department of Infectious Disease Medicine, The First Hospital Affiliated to Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
10
|
Wu S, Wang X, Xing W, Li F, Liang M, Li K, He Y, Wang J. An update on animal models of liver fibrosis. Front Med (Lausanne) 2023; 10:1160053. [PMID: 37035335 PMCID: PMC10076546 DOI: 10.3389/fmed.2023.1160053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The development of liver fibrosis primarily determines quality of life as well as prognosis. Animal models are often used to model and understand the underlying mechanisms of human disease. Although organoids can be used to simulate organ development and disease, the technology still faces significant challenges. Therefore animal models are still irreplaceable at this stage. Currently, in vivo models of liver fibrosis can be classified into five categories based on etiology: chemical, dietary, surgical, transgenic, and immune. There is a wide variety of animal models of liver fibrosis with varying efficacy, which have different implications for proper understanding of the disease and effective screening of therapeutic agents. There is no high-quality literature recommending the most appropriate animal models. In this paper, we will describe the progress of commonly used animal models of liver fibrosis in terms of their development mechanisms, applications, advantages and disadvantages, and recommend appropriate animal models for different research purposes.
Collapse
Affiliation(s)
- ShuTing Wu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - WenBo Xing
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - FenYao Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Ming Liang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - KeShen Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Yan He,
| | - JianMing Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Department of Hepatobiliary and Pancreatic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- JianMing Wang,
| |
Collapse
|