1
|
Zhao L, Tang H, Cheng Z. Pharmacotherapy of Liver Fibrosis and Hepatitis: Recent Advances. Pharmaceuticals (Basel) 2024; 17:1724. [PMID: 39770566 PMCID: PMC11677259 DOI: 10.3390/ph17121724] [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: 11/10/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a progressive scarring process primarily caused by chronic inflammation and injury, often closely associated with viral hepatitis, alcoholic liver disease, metabolic dysfunction-associated steatotic liver disease (MASLD), drug-induced liver injury, and autoimmune liver disease (AILD). Currently, there are very few clinical antifibrotic drugs available, and effective targeted therapy is lacking. Recently, emerging antifibrotic drugs and immunomodulators have shown promising results in animal studies, and some have entered clinical research phases. This review aims to systematically review the molecular mechanisms underlying liver fibrosis, focusing on advancements in drug treatments for hepatic fibrosis. Furthermore, since liver fibrosis is a progression or endpoint of many diseases, it is crucial to address the etiological treatment and secondary prevention for liver fibrosis. We will also review the pharmacological treatments available for common hepatitis leading to liver fibrosis.
Collapse
Affiliation(s)
- Liangtao Zhao
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Haolan Tang
- School of Medicine, Southeast University, Nanjing 210009, China;
| | - Zhangjun Cheng
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| |
Collapse
|
2
|
Ciceu A, Fenyvesi F, Hermenean A, Ardelean S, Dumitra S, Puticiu M. Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems. Int J Mol Sci 2024; 25:9346. [PMID: 39273295 PMCID: PMC11394827 DOI: 10.3390/ijms25179346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Chronic liver injuries often lead to hepatic fibrosis, a condition characterized by excessive extracellular matrix accumulation and abnormal connective tissue hyperplasia. Without effective treatment, hepatic fibrosis can progress to cirrhosis or hepatocellular carcinoma. Current treatments, including liver transplantation, are limited by donor shortages and high costs. As such, there is an urgent need for effective therapeutic strategies. This review focuses on the potential of plant-based therapeutics, particularly polyphenols, phenolic acids, and flavonoids, in treating hepatic fibrosis. These compounds have demonstrated anti-fibrotic activities through various signaling pathways, including TGF-β/Smad, AMPK/mTOR, Wnt/β-catenin, NF-κB, PI3K/AKT/mTOR, and hedgehog pathways. Additionally, this review highlights the advancements in nanoparticulate drug delivery systems that enhance the pharmacokinetics, bioavailability, and therapeutic efficacy of these bioactive compounds. Methodologically, this review synthesizes findings from recent studies, providing a comprehensive analysis of the mechanisms and benefits of these plant-based treatments. The integration of novel drug delivery systems with plant-based therapeutics holds significant promise for developing effective treatments for hepatic fibrosis.
Collapse
Affiliation(s)
- Alina Ciceu
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Ferenc Fenyvesi
- Department of Molecular and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Simona Ardelean
- Faculty of Pharmacy, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Simona Dumitra
- Faculty of Medicine, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Monica Puticiu
- Faculty of Medicine, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| |
Collapse
|
3
|
Musso G, Pinach S, Mariano F, Saba F, De Michieli F, Framarin L, Berrutti M, Paschetta E, Parente R, Lizet Castillo Y, Leone N, Castellino F, Cassader M, Gambino R. Effect of phospholipid curcumin Meriva on liver histology and kidney disease in nonalcoholic steatohepatitis: A randomized, double-blind, placebo-controlled trial. Hepatology 2024:01515467-990000000-00891. [PMID: 38809154 DOI: 10.1097/hep.0000000000000937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/14/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND AND AIMS NASH confers an increased liver-related and kidney morbidity. Phospholipid curcumin (Meriva) is a phospholipid formulation with ameliorated systemic curcumin absorption and delivery. We assessed the safety and efficacy of Meriva in NASH. APPROACH AND RESULTS In this double-blind trial, 52 patients with biopsy-proven NASH (71% with stage ≥F2 fibrosis, 58% with stage A2-G2/A2-G3a chronic kidney disease) were randomized 1:1 to receive Meriva 2 g/d or placebo for 72 weeks. The primary endpoint was NASH resolution with no worsening of fibrosis. The secondary endpoints included a ≥1 stage liver fibrosis improvement with no NASH worsening; regression of significant (ie, stage ≥F2) fibrosis and CKD; and improvement in renal, glucose, lipid, and inflammatory parameters. We also explored the treatment effect on hepatic activation of NF-kB, a key proinflammatory transcription factor and a major target of curcumin. Fifty-one patients (26 on Meriva and 25 on placebo) completed the trial. Sixteen (62%) patients on Meriva versus 3 (12%) patients on placebo had NASH resolution (RR = 5.33 [95% CI = 1.76-12.13]; p = 0.003). Thirteen (50%) patients on Meriva versus 2 (8%) patients on placebo had ≥1 stage fibrosis improvement (RR = 6.50 [1.63-21.20]; p = 0.008). Eleven (42%) patients on Meriva versus 0 (0%) on placebo had regression of significant liver fibrosis (RR = 18.01 [1.43-36.07]; p = 0.02). Hepatic NF-kB inhibition predicted NASH resolution (AUC = 0.90, 95% CI = 0.84-0.95) and fibrosis improvement (AUC = 0.89, 95% CI = 0.82-0.96). Thirteen (50%) patients on Meriva versus 0 (0%) on placebo had chronic kidney disease regression (RR = 10.71 [1.94-17.99)]; p = 0.004). Compared with placebo, Meriva improved eGFR (difference in adjusted eGFR change: +3.59 [2.96-4.11] mL/min/1.73 m 2 /y, p = 0.009), fasting glucose(-17 mg/dL; 95% CI = -22, -12), HbA1c (-0.62%; 95% CI = -0.87%, -0.37%), LDL-C (-39 mg/dL; 95% CI = -45, -33), triglycerides (-36 mg/dL, 95% CI = -46, -26), HDL-C (+10 mg/dL; 95% CI = +8, +11), and inflammatory markers. Adverse events were rare, mild, and evenly distributed. CONCLUSIONS In patients with NASH, Meriva administration for 72 weeks was safe, well-tolerated, and improved liver histology, possibly through NF-kB inhibition, kidney disease, and metabolic profile.
Collapse
Affiliation(s)
- Giovanni Musso
- MECAU Department, San Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Silvia Pinach
- Department of Medical Sciences, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Filippo Mariano
- Department of Nephrology, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Francesca Saba
- Department of Medical Sciences, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Franco De Michieli
- Department of Medical Sciences, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Luciana Framarin
- Gastroenterology Unit, HUMANITAS Gradenigo Hospital, Turin, Italy
| | - Mara Berrutti
- Gastroenterology Unit, HUMANITAS Gradenigo Hospital, Turin, Italy
| | - Elena Paschetta
- MECAU Department, HUMANITAS Gradenigo Hospital, Turin, Italy
| | - Renato Parente
- Pathology Unit, HUMANITAS Gradenigo Hospital, Turin, Italy
| | | | - Nicola Leone
- Gastroenterology Unit, HUMANITAS Gradenigo Hospital, Turin, Italy
| | | | - Maurizio Cassader
- Department of Medical Sciences, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Vanzan DF, Goma EP, Locatelli FR, Honorio TDS, Furtado PDS, Rodrigues CR, de Sousa VP, Mata dos Santos HA, do Carmo FA, Simon A, Pyrrho ADS, Ribeiro AJ, Cabral LM. Evaluation of Silybin Nanoparticles against Liver Damage in Murine Schistosomiasis mansoni Infection. Pharmaceutics 2024; 16:618. [PMID: 38794280 PMCID: PMC11125168 DOI: 10.3390/pharmaceutics16050618] [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: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Silybin (SIB) is a hepatoprotective drug known for its poor oral bioavailability, attributed to its classification as a class IV drug with significant metabolism during the first-pass effect. This study explored the potential of solid lipid nanoparticles with (SLN-SIB-U) or without (SLN-SIB) ursodeoxycholic acid and polymeric nanoparticles (PN-SIB) as delivery systems for SIB. The efficacy of these nanosystems was assessed through in vitro studies using the GRX and Caco-2 cell lines for permeability and proliferation assays, respectively, as well as in vivo experiments employing a murine model of Schistosomiasis mansoni infection in BALB/c mice. The mean diameter and encapsulation efficiency of the nanosystems were as follows: SLN-SIB (252.8 ± 4.4 nm, 90.28 ± 2.2%), SLN-SIB-U (252.9 ± 14.4 nm, 77.05 ± 2.8%), and PN-SIB (241.8 ± 4.1 nm, 98.0 ± 0.2%). In the proliferation assay with the GRX cell line, SLN-SIB and SLN-SIB-U exhibited inhibitory effects of 43.09 ± 5.74% and 38.78 ± 3.78%, respectively, compared to PN-SIB, which showed no inhibitory effect. Moreover, SLN-SIB-U demonstrated a greater apparent permeability coefficient (25.82 ± 2.2) than PN-SIB (20.76 ± 0.1), which was twice as high as that of SLN-SIB (11.32 ± 4.6) and pure SIB (11.28 ± 0.2). These findings suggest that solid lipid nanosystems hold promise for further in vivo investigations. In the murine model of acute-phase Schistosomiasis mansoni infection, both SLN-SIB and SLN-SIB-U displayed hepatoprotective effects, as evidenced by lower alanine amino transferase values (22.89 ± 1.6 and 23.93 ± 2.4 U/L, respectively) than those in control groups I (29.55 ± 0.7 U/L) and I+SIB (34.29 ± 0.3 U/L). Among the prepared nanosystems, SLN-SIB-U emerges as a promising candidate for enhancing the pharmacokinetic properties of SIB.
Collapse
Affiliation(s)
- Daniel Figueiredo Vanzan
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ester Puna Goma
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Fernanda Resende Locatelli
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Thiago da Silva Honorio
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Priscila de Souza Furtado
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Carlos Rangel Rodrigues
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Valeria Pereira de Sousa
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Hilton Antônio Mata dos Santos
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Flávia Almada do Carmo
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Alice Simon
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Alexandre dos Santos Pyrrho
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - António José Ribeiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- Group Genetics of Cognitive Dysfunction, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4169-007 Porto, Portugal
| | - Lucio Mendes Cabral
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| |
Collapse
|
5
|
Du YQ, Yuan B, Ye YX, Zhou FL, Liu H, Huang JJ, Wei YF. Plumbagin Regulates Snail to Inhibit Hepatocellular Carcinoma Epithelial-Mesenchymal Transition in vivo and in vitro. J Hepatocell Carcinoma 2024; 11:565-580. [PMID: 38525157 PMCID: PMC10960549 DOI: 10.2147/jhc.s452924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/01/2024] [Indexed: 03/26/2024] Open
Abstract
Background/Aims Plumbagin (PL) has been shown to effe ctively inhibit autophagy, suppressing invasion and migration of hepatocellular carcinoma (HCC) cells. However, the specific mechanism remains unclear. This study aimed to investigate the effect of PL on tumor growth factor (TGF)-β-induced epithelial-mesenchymal transition (EMT) in HCC. Methods Huh-7 cells were cultured, and in vivo models of EMT and HCC-associated lung metastasis were developed through tail vein and in situ injections of tumor cells. In vivo imaging and hematoxylin and eosin staining were used to evaluate HCC modeling and lung metastasis. After PL intervention, the expression levels of Snail, vimentin, E-cadherin, and N-cadherin in the liver were evaluated through immunohistochemistry and Western blot. An in vitro TGF-β-induced cell EMT model was used to detect Snail, vimentin, E-cadherin, and N-cadherin mRNA levels through a polymerase chain reaction. Their protein levels were detected by immunofluorescence staining and Western blot. Results In vivo experiments demonstrated that PL significantly reduced the expression of Snail, vimentin, and N-cadherin, while increasing the expression of E-cadherin at the protein levels, effectively inhibiting HCC and lung metastasis. In vitro experiments confirmed that PL up-regulated epithelial cell markers, down-regulated mesenchymal cell markers, and inhibited EMT levels in HCC cells. Conclusion PL inhibits Snail expression, up-regulates E-cadherin expression, and down-regulates N-cadherin and vimentin expression, preventing EMT in HCC cells and reducing lung metastasis.
Collapse
Affiliation(s)
- Yuan-Qin Du
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Bin Yuan
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Yi-Xian Ye
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Feng-ling Zhou
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Hong Liu
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Jing-Jing Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530024, People’s Republic of China
| | - Yan-Fei Wei
- Department of Physiology, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, 530200, People’s Republic of China
| |
Collapse
|
6
|
Zein N, Yassin F, Ayoub HG, Elewa YHA, Mohamed SKA, Mahmoud MH, Elfeky M, Batiha GES, Zahran MH. In vivo investigation of the anti-liver fibrosis impact of Balanites aegyptiaca/ chitosan nanoparticles. Biomed Pharmacother 2024; 172:116193. [PMID: 38301419 DOI: 10.1016/j.biopha.2024.116193] [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: 10/06/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024] Open
Abstract
Balanites aegyptiaca (B. aegyptiaca) is an African herb with traditional medical applications. Various pathogenic factors cause hepatic fibrosis and require novel treatment alternatives. Nanoformulation-based natural products can overcome the available drug problems by increasing the efficacy of natural products targeting disease markers. The current study investigated B. aegyptiaca methanolic extract using high-pressure liquid chromatography (HPLC), and B. aegyptiaca/chitosan nanoparticles were prepared. In vivo, evaluation tests were performed to assess the curative effect of the successfully prepared B. aegyptiaca/chitosan nanoparticles. For 30 days, the rats were divided into six groups, typical and fibrosis groups, where the liver fibrosis groups received B. aegyptiaca extract, silymarin, chitosan nanoparticles, and B. aegyptiaca/chitosan nanoparticles daily. In the current investigation, phenolic molecules are the major compounds detected in B. aegyptiaca extract. UV showed that the prepared B. aegyptiaca /chitosan nanoparticles had a single peak at 280 nm, a particle size of 35.0 ± 6.0 nm, and a negative charge at - 8.3 mV. The animal studies showed that the synthetic B. aegyptiaca/chitosan nanoparticles showed substantial anti-fibrotic protective effects against CCl4-induced hepatic fibrosis in rats when compared with other groups through optimization of biochemical and oxidative markers, improved histological changes, and modulated the expression of Col1a1, Acta2 and Cxcl9 genes, which manage liver fibrosis. In conclusion, the current research indicated that the prepared B. aegyptiaca/chitosan nanoparticles improved histological structure and significantly enhanced the biochemical and genetic markers of liver fibrosis in an animal model.
Collapse
Affiliation(s)
- Nabila Zein
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Fathy Yassin
- Chemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Heba G Ayoub
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Sherif Kh A Mohamed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Kingdom of Saudi Arabia
| | - Mohamed Elfeky
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur 22511, AlBeheira, Egypt
| | - Mahmoud Hosny Zahran
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
7
|
Zhao Y, Dai E, Dong L, Yuan J, Zhao Y, Wu T, Kong R, Li M, Wang S, Zhou L, Yang Y, Kong H, Zhao Y, Qu H. Available and novel plant-based carbon dots derived from Vaccaria Semen carbonisata alleviates liver fibrosis. Front Mol Biosci 2023; 10:1282929. [PMID: 38116381 PMCID: PMC10729316 DOI: 10.3389/fmolb.2023.1282929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/27/2023] [Indexed: 12/21/2023] Open
Abstract
Background: Liver fibrosis represents an intermediate stage in the progression of liver disease, and as of now, there exists no established clinical therapy for effective antifibrotic treatment. Purpose: Our aim is to explore the impact of Carbon dots derived from Vaccaria Semen Carbonisata (VSC-CDs) on carbon tetrachloride-induced liver fibrosis in mice. Methods: VSC-CDs were synthesized employing a modified pyrolysis process. Comprehensive characterization was performed utilizing various techniques, including transmission electron microscopy (TEM), multiple spectroscopies, X-ray photoelectron spectroscopy (XPS), and high-performance liquid chromatography (HPLC). A hepatic fibrosis model induced by carbon tetrachloride was utilized to evaluate the anti-hepatic fibrosis effects of VSC-CDs. Results: VSC-CDs, exhibiting a quantum yield (QY) of approximately 2.08%, were nearly spherical with diameters ranging from 1.0 to 5.5 nm. The VSC-CDs prepared in this study featured a negative charge and abundant chemical functional groups. Furthermore, these particles demonstrated outstanding dispersibility in the aqueous phase and high biocompatibility. Moreover, VSC-CDs not only enhanced liver function and alleviated liver damage in pathomorphology but also mitigated the extent of liver fibrosis. Additionally, this study marks the inaugural demonstration of the pronounced activity of VSC-CDs in inhibiting inflammatory reactions, reducing oxidative damage, and modulating the TGF-β/Smad signaling pathway. Conclusion: VSC-CDs exerted significant potential for application in nanodrugs aimed at treating liver fibrosis.
Collapse
Affiliation(s)
- Yafang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ertong Dai
- Qingdao Eighth People’s Hospital, Qingdao, Shandong, China
| | - Liyang Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jinye Yuan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yusheng Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tong Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ruolan Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Menghan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuxian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Long Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yingxin Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huihua Qu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Sun A, Liu H, Sun M, Yang W, Liu J, Lin Y, Shi X, Sun J, Liu L. Emerging nanotherapeutic strategies targeting gut-X axis against diseases. Biomed Pharmacother 2023; 167:115577. [PMID: 37757494 DOI: 10.1016/j.biopha.2023.115577] [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: 08/08/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023] Open
Abstract
Gut microbiota can coordinate with different tissues and organs to maintain human health, which derives the concept of the gut-X axis. Conversely, the dysbiosis of gut microbiota leads to the occurrence and development of various diseases, such as neurological diseases, liver diseases, and even cancers. Therefore, the modulation of gut microbiota offers new opportunities in the field of medicines. Antibiotics, probiotics or other treatments might restore unbalanced gut microbiota, which effects do not match what people have expected. Recently, nanomedicines with the high targeting ability and reduced toxicity make them an appreciative choice for relieving disease through targeting gut-X axis. Considering this paradigm-setting trend, the current review summarizes the advancements in gut microbiota and its related nanomedicines. Specifically, this article introduces the immunological effects of gut microbiota, summarizes the gut-X axis-associated diseases, and highlights the nanotherapeutics-mediated treatment via remolding the gut-X axis. Moreover, this review also discusses the challenges in studies related to nanomedicines targeting the gut microbiota and offers the future perspective, thereby aiming at charting a course toward clinic.
Collapse
Affiliation(s)
- Ao Sun
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hongyu Liu
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, Liaoning Province, China; Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, China Medical University, Ministry of Education, Shenyang, Liaoning Province, China
| | - Mengchi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, PR China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jiaxin Liu
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yi Lin
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, PR China.
| | - Linlin Liu
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
9
|
Filip M, Baldea I, David L, Moldovan B, Flontas GC, Macavei S, Muntean DM, Decea N, Tigu AB, Clichici SV. Hybrid Material Based on Vaccinium myrtillus L. Extract and Gold Nanoparticles Reduces Oxidative Stress and Inflammation in Hepatic Stellate Cells Exposed to TGF-β. Biomolecules 2023; 13:1271. [PMID: 37627336 PMCID: PMC10452749 DOI: 10.3390/biom13081271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: The study aimed to investigate the impact of gold nanoparticles capped with Cornus sanguinea (NPCS) and mixed with a fruit extract (Vaccinum myrtillus L.-VL) on human hepatic stellate cells (LX-2) exposed to TGF-β. (2) Methods: NPCS were characterized by UV-Vis, transmission electron microscopy (TEM), zeta potential measurement, X-ray diffraction (XRD) and energy dispersive spectroscopy (EDX). The cytotoxic effects of VL, NPCS and of the hybrid compounds obtained by mixing the two components in variable proportions (NPCS-VL) were assessed. LDH activity, MDA levels, secretion of inflammation markers, the expression of fibrogenesis markers and collagen I synthesis were estimated after treating the cells with a mixture of 25:25 μg/mL NPCS and VL. (3) Results: TEM analysis showed that NPCS had spherical morphology and homogenous distribution, while their formation and elemental composition were confirmed by XRD and EDX analysis. TGF-β increased cell membrane damage as well as secretion of IL-1β, IL-1α and TLR4. It also amplified the expression of α-SMA and type III collagen and induced collagen I deposition. NPCS administration reduced the inflammation caused by TGF-β and downregulated α-SMA expression. VL diminished LDH activity and the secretion of proinflammatory cytokines. The NPCS-VL mixture maintained IL-1β, IL-1α, TLR4 and LDH at low levels after TGF-β exposure, but it enhanced collagen III expression. (4) Conclusions: The mixture of NPCS and VL improved cell membrane damage and inflammation triggered by TGF-β and mitigated collagen I deposition, but it increased the expression of collagen III, suggestive of a fibrogenetic effect of the hybrid material.
Collapse
Affiliation(s)
- Mara Filip
- Department of Physiology, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 1–3 Clinicilor Street, 400006 Cluj-Napoca, Romania; (G.C.F.); (N.D.); (S.V.C.)
| | - Ioana Baldea
- Department of Physiology, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 1–3 Clinicilor Street, 400006 Cluj-Napoca, Romania; (G.C.F.); (N.D.); (S.V.C.)
| | - Luminita David
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering “Babes-Bolyai” University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania; (L.D.); (B.M.)
| | - Bianca Moldovan
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering “Babes-Bolyai” University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania; (L.D.); (B.M.)
| | - Gabriel Cristian Flontas
- Department of Physiology, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 1–3 Clinicilor Street, 400006 Cluj-Napoca, Romania; (G.C.F.); (N.D.); (S.V.C.)
| | - Sergiu Macavei
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donath Street, No. 67-103, 400293 Cluj-Napoca, Romania;
| | - Dana Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 8 Victor Babeș Street, 400347 Cluj-Napoca, Romania;
| | - Nicoleta Decea
- Department of Physiology, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 1–3 Clinicilor Street, 400006 Cluj-Napoca, Romania; (G.C.F.); (N.D.); (S.V.C.)
| | - Adrian Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4 Louis Pasteur Street, 400347 Cluj-Napoca, Romania;
| | - Simona Valeria Clichici
- Department of Physiology, ‘‘Iuliu Hatieganu’’ University of Medicine and Pharmacy, 1–3 Clinicilor Street, 400006 Cluj-Napoca, Romania; (G.C.F.); (N.D.); (S.V.C.)
| |
Collapse
|