Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Toxicol Mech Methods ; : 1-8, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38685856

ABSTRACT

Liver fibrosis is a common pathological process in the progression of several chronic liver diseases to cirrhosis and hepatocellular carcinoma. Therefore, the development of medications that can repress the progress of liver fibrosis is essential. We discovered that initially, 12ß-(m-methyl-benzoyl)-11,12-dihydro oleanolic acid (12d-OA), a farnesoid X receptor (FXR) modulator, possessed potential anti-fibrotic properties. Through an in-depth study, we revealed that 12d-OA not only inhibited the expression of fibrogenic markers in the LX-2 cells and HSC-T6 cells but also exhibited significant protective effects against liver injury and liver fibrosis in bile duct ligation (BDL) rats. Further exploration of its molecular mechanism indicated that 12d-OA exerted antifibrotic activity by inhibiting the extracellular signal-regulated kinase (ERK)/stress-activated protein kinase (p38) signaling pathways. Consequently, the great effects of 12d-OA in vitro and in vivo suggest that it may be a good candidate for liver fibrosis.

2.
J Med Chem ; 66(12): 7969-7987, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37294950

ABSTRACT

A series of new tricyclic matrinane derivatives were continuously synthesized and evaluated for their inhibitory effects on genes and proteins related to hepatic fibrosis at the cellular level, including collagen type I α1 chain (COL1A1), α smooth muscle actin (α-SMA), connective tissue growth factor (CTGF), and matrix metalloprotein 2 (MMP-2). Among them, compound 6k exerted an appealing potency and significantly reduced liver injury and fibrosis in both bile duct ligation (BDL) rats and Mdr2 knockout mice. An activity-based protein profiling (ABPP) assay indicated that 6k might directly bind to Ewing sarcoma breakpoint region 1 (EWSR1) to inhibit its function and affect the expression of downstream liver fibrosis-related genes and thus regulate liver fibrosis. These results provided a potential novel target for the treatment of liver fibrosis and powerful information for the development of tricyclic matrinanes into promising anti-hepatic fibrosis agents.


Subject(s)
Matrines , Sarcoma, Ewing , Animals , Mice , Rats , Antifibrotic Agents , Fibrosis , Liver , Liver Cirrhosis/pathology , Sarcoma, Ewing/pathology , RNA-Binding Protein EWS
3.
Pharmaceuticals (Basel) ; 16(5)2023 May 17.
Article in English | MEDLINE | ID: mdl-37242541

ABSTRACT

The farnesoid X receptor (FXR) plays a crucial role in regulating the metabolism of bile acids, lipids, and sugars. Consequently, it is implicated in the treatment of various diseases, including cholestasis, diabetes, hyperlipidemia, and cancer. The advancement of novel FXR modulators holds immense importance, especially in managing metabolic disorders. In this study, a series of oleanolic acid (OA) derivatives bearing 12ß-O-(γ-glutamyl) groups were designed and synthesized. Using a yeast one-hybrid assay, we established a preliminary structure-activity relationship (SAR) and identified the most potent compound, 10b, which selectively antagonizes FXR over other nuclear receptors. Compound 10b can differentially modulate the downstream genes of FXR, including with the upregulation of the CYP7A1 gene. In vivo testing revealed that 10b (100 mg·Kg-1) not only effectively inhibits lipid accumulation in the liver but also prevents liver fibrosis in both BDL rats and HFD mice. Molecular modeling indicated that the branched substitution of 10b extends into the H11-H12 region of FXR-LBD, possibly accounting for its CYP7A1 upregulation, which is different from a known OA 12ß-alkonate. These findings suggest that 12-glutamyl OA derivative 10b represents a promising candidate for the treatment of nonalcoholic steatohepatitis (NASH).

4.
Phytother Res ; 37(9): 3898-3912, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37132081

ABSTRACT

Liver fibrosis is a key global health care burden. Sclareol, isolated from Salvia sclarea, possesses various biological activities. Its effect on liver fibrosis remains unknown. This study was proposed to evaluate the antifibrotic activity of sclareol (SCL) and explore its underlying mechanisms. Stimulated hepatic stellate cells served as an in vitro liver fibrosis model. The expression of fibrotic markers was assessed by western blot and real-time PCR. Two classical animal models, bile duct-ligated rats and carbon tetrachloride-treated mice, were utilized for the in vivo experiments. The liver function and fibrosis degree were determined by serum biochemical and histopathological analyses. VEGFR2 SUMOylation was analyzed using coimmunoprecipitation assay. Our results indicated that SCL treatment restricted the profibrotic propensity of activated HSCs. In fibrotic rodents, SCL administration alleviated hepatic injury and reduced collagen accumulation. Mechanistic studies indicated that SCL downregulated the protein level of SENP1 and enhanced VEGFR2 SUMOylation in LX-2 cells, which affected its intracellular trafficking. Blockade of the interaction between VEGFR2 and STAT3 was observed, resulting in the suppression of downstream STAT3 phosphorylation. Our findings demonstrated that SCL has therapeutic efficacy against liver fibrosis through mediating VEGFR2 SUMOylation, suggesting that SCL may be a potential candidate compound for its treatment.


Subject(s)
Liver Cirrhosis , Sumoylation , Rats , Mice , Animals , Liver Cirrhosis/drug therapy , Liver , Signal Transduction , Fibrosis , Hepatic Stellate Cells
5.
Bioorg Med Chem Lett ; 88: 129290, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37080476

ABSTRACT

A novel benzene sulfonamide compound named IMB16-4 exhibits excellent anti-hepatic fibrosis activity in a recent study. To develop potential anti-hepatic fibrosis agents, a series of benzene sulfonamide derivatives were designed and synthesized based on the scaffold of the lead compound IMB16-4. As it turned out, most of the derivatives displayed potential anti-hepatic fibrosis activity, among which, compounds 11a, 11b, 11d, 13a, 36b, and 47b exhibited inhibition rates of 42.3%, 48.7%, 42.4%, 40.0%, 39.4%, and 49.3%, respectively, which were equivalent to the control IMB16-4 with an inhibition rate of 35.9%, Costunolide with an inhibition rate of 45.4%, and much more potent than that of Epigallocatechin gallate (EGCG) with an inhibition rate of 25.3%. Especially, compounds 46a, 46b, and 46c exhibited excellent anti-hepatic fibrosis activity with inhibition rates of 61.7%, 54.8%, and 60.7%, which were almost 1.5-fold inhibition rates of IMB16-4. In addition, compounds 46a, 46b, and 46c exhibited remarkable inhibitory activity in the gene expression of COL1A1, MMP-2, and the protein expression of COL1A1, FN, α-SMA, and TIMP-1 by inhibiting the JAK1-STAT1/3 pathway. These findings furnished valuable inspiration for the further development of anti-hepatic fibrosis agents.


Subject(s)
Antifibrotic Agents , Benzene , Humans , Benzene Derivatives , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Sulfonamides/pharmacology , Structure-Activity Relationship
6.
Eur J Med Chem ; 245(Pt 1): 114886, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36347091

ABSTRACT

Sixty-one palmatine (PMT) derivatives, of which twenty-eight were new, were synthesized and evaluated for their anti-fibrogenic activities via collagen type I α 1 (COL1A1)-promoter based luciferase model in LX-2 cells, taking 2,3,10-trimethoxy-9-p-isopropyloxyprotopalmatine bromide (1) as the lead. Among them, compound 3a exerted the highest potency with the IC50 value of 8.19 µmol/L and SI value of 8.59, and reduced the expressions of multiple fibrogenic biomarkers, including COL1A1, TGF-ß1, α-SMA and TIMP1 in a dose-dependent manner. In addition, it significantly reduced liver steatosis and inflammation, and especially attenuated the degree of liver fibrosis in choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD)-induced NASH mice model in vivo. Mechanism study indicated that it significantly ameliorated liver injury by activating farnesoid X receptor (FXR). BDL-induced fibrosis rats model further verified its liver-protective and anti-fibrosis activities. Therefore, PMT derivatives constituted a new family of non-steroidal FXR agonists as anti-NASH candidates, with the advantage of good safety profile, and are worthy for further investigation.


Subject(s)
Antifibrotic Agents , Berberine Alkaloids , Liver , Non-alcoholic Fatty Liver Disease , Animals , Mice , Rats , Berberine Alkaloids/chemistry , Berberine Alkaloids/pharmacology , Berberine Alkaloids/therapeutic use , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Antifibrotic Agents/chemistry , Antifibrotic Agents/pharmacology , Antifibrotic Agents/therapeutic use
7.
Bioorg Chem ; 127: 105971, 2022 10.
Article in English | MEDLINE | ID: mdl-35749855

ABSTRACT

Liver fibrosis is an important process in chronic liver disease and is strongly related to poor prognosis. Dehydromevalonolactone (C8) is a natural product isolated from a fungus of Fusarium sp. CPCC 401218, and its pharmacological activity has never been reported before. In this study, the potential of C8 as an anti-hepatic fibrosis agent was investigated. In human hepatic stellate cell (HSC) line LX-2, C8 suppressed the increased expression of COL1A1 and α-SMA induced by TGFß1, which indicated that C8 could repress the activation of HSCs. In bile duct ligated rats, C8 administration (100 mg/kg, i.p.) markedly attenuated liver injury, fibrosis, and inflammation, and suppressed the expression of the macrophage surface marker F4/80. In terms of mechanism, C8 treatment blocked the activation of the NLRP3 inflammasome, which was stimulated by LPS and nigericin in bone marrow-derived macrophages (BMDMs) and companied by the release of active IL-1ß. In addition, the activation of LX-2 cells induced by IL-1ß released from BMDMs was also inhibited after C8 administration, which indicated that C8 repressed HSCs activation by inhibiting the activation of NLRP3 inflammasome in macrophages. Furthermore, C8 exhibited the effects of anti-fibrosis and inhibiting the expression of NLRP3 inflammasome in non-alcoholic steatohepatitis (NASH) mice. Finally, C8 can be commendably absorbed in vivo and was safe for mice at the concentration of 1000 mg/kg (p.o.). In summary, our study reveals that C8 ameliorates HSCs activation and liver fibrosis in cholestasis rats and NASH mice by inhibiting NLRP3 inflammasome in macrophages, and C8 might be a safe and effective candidate for the treatment of liver fibrosis.


Subject(s)
Inflammasomes , Mevalonic Acid/analogs & derivatives , Non-alcoholic Fatty Liver Disease , Animals , Fibrosis , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Mevalonic Acid/analysis , Mevalonic Acid/pharmacology , Mevalonic Acid/therapeutic use , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Rats
8.
Acta Pharmacol Sin ; 42(11): 1808-1820, 2021 11.
Article in English | MEDLINE | ID: mdl-34465912

ABSTRACT

Liver fibrosis is one of the most severe pathologic consequences of chronic liver diseases, and effective therapeutic strategies are urgently needed. Proton pump inhibitors (PPIs) are H+/K+-ATPase inhibitors and currently used to treat acid-related diseases such as gastric ulcers, which have shown other therapeutic effects in addition to inhibiting acid secretion. However, few studies have focused on PPIs from the perspective of inhibiting hepatic fibrosis. In the present study, we investigated the effects of pantoprazole (PPZ), a PPI, against liver fibrosis in a bile duct ligation (BDL) rat model, human hepatic stellate cell (HSC) line LX-2 and mouse primary HSCs (pHSCs), and explored the potential mechanisms underlying the effects of PPZ in vitro and in vivo. In BDL rats, administration of PPZ (150 mg· kg-1· d-1, i.p. for 14 d) significantly attenuated liver histopathological injury, collagen accumulation, and inflammatory responses, and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, Tgfß1, and Mmp-2. In LX-2 cells and mouse pHSCs, PPZ (100-300 µM) dose-dependently suppressed the levels of fibrogenic markers. We conducted transcriptome analysis and subsequent validation in PPZ-treated LX-2 cells, and revealed that PPZ inhibited the expression of Yes-associated protein (YAP) and its downstream targets such as CTGF, ID1, survivin, CYR61, and GLI2. Using YAP overexpression and silencing, we demonstrated that PPZ downregulated hepatic fibrogenic gene expression via YAP. Furthermore, we showed that PPZ promoted the proteasome-dependent degradation and ubiquitination of YAP, thus inhibiting HSC activation. Additionally, we showed that PPZ destabilized YAP by disrupting the interaction between a deubiquitinating enzyme OTUB2 and YAP, and subsequently blocked the progression of hepatic fibrosis.


Subject(s)
Bile Ducts/drug effects , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Pantoprazole/therapeutic use , Proteolysis/drug effects , YAP-Signaling Proteins/agonists , Animals , Bile Ducts/metabolism , Gene Expression Profiling , HEK293 Cells , Hepatic Stellate Cells/metabolism , Humans , Ligation , Liver Cirrhosis/metabolism , Male , Pantoprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , YAP-Signaling Proteins/metabolism
9.
Molecules ; 25(21)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121156

ABSTRACT

Twenty-seven novel 12N-substituted aloperine derivatives were synthesized and investigated for their inhibitory effects on collagen α1 (I) (COL1A1) promotor in human hepatic stellate LX-2 cells, taking aloperine (1) as the hit. A structure-activity relationship (SAR) study disclosed that the introduction of suitable substituents on the 12N atom might enhance the activity. Compound 4p exhibited a good promise on down-regulating COL1A1 expression with the IC50 value of 16.5 µM. Its inhibitory activity against COL1A1 was further confirmed on both mRNA and protein levels. Meanwhile, it effectively inhibited the expression of other fibrogenic proteins, such as transforming growth factor ß1 (TGF-ß1) and smooth muscle actin (α-SMA). It also exhibited good in vivo safety profile with the oral LD50 value of 400 mg kg-1 in mice. The results initiated the anti-liver fibrogenic study of aloperine derivatives, and the key compound 4p was selected as a novel lead for further investigation against liver fibrogenesis.


Subject(s)
Liver/drug effects , Liver/pathology , Piperidines/chemistry , Piperidines/pharmacology , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cytoprotection/drug effects , Drug Design , Fibrosis , Gene Expression Regulation/drug effects , Humans , Liver/metabolism , Piperidines/adverse effects , Promoter Regions, Genetic/genetics , Quinolizidines , Safety , Structure-Activity Relationship , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
10.
Br J Pharmacol ; 177(2): 372-387, 2020 01.
Article in English | MEDLINE | ID: mdl-31621893

ABSTRACT

BACKGROUND AND PURPOSE: This study investigates the antifibrotic activities and potential mechanisms of costunolide (COS), a natural sesquiterpene compound. EXPERIMENTAL APPROACH: Rats subjected to bile duct ligation and mice challenged with CCl4 were used to study the antifibrotic effects of COS in vivo. Mouse primary hepatic stellate cells (pHSCs) and human HSC line LX-2 also served as an in vitro liver fibrosis models. The expression of fibrogenic genes and signaling proteins in the neurogenic locus notch homologue protein 3 (Notch3)-hairy/enhancer of split-1 (HES1) pathway was examined using western blot and/or real-time PCR. Notch3 degradation was analysed using immunofluorescence and coimmunoprecipitation. KEY RESULTS: In animals, COS administration attenuated hepatic histopathological injury and collagen accumulation and reduced the expression of fibrogenic genes. COS time- and dose-dependently suppressed the levels of fibrotic markers in LX-2 cells and mouse pHSCs. Mechanistic studies showed COS destabilized Notch3 and subsequently inhibited the Notch3-HES1 pathway, thus inhibiting HSC activation. Furthermore, COS blocked the WW domain-containing protein 2 (WWP2)/protein phosphatase 1G (PPM1G) interaction and enhanced the effect of WWP2 on Notch3 degradation. CONCLUSIONS AND IMPLICATIONS: COS exerted potent antifibrotic effects in vitro and in vivo by disrupting the WWP2/PPM1G complex, promoting Notch3 degradation and inhibiting the Notch3/HES1 pathway. This indicates that COS may be a potential therapeutic candidate for the treatment of liver fibrosis.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Receptor, Notch3/metabolism , Sesquiterpenes/pharmacology , Ubiquitin-Protein Ligases/metabolism , Animals , Carbon Tetrachloride , Cell Line , Common Bile Duct/surgery , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligation , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred BALB C , Proteolysis , Rats, Sprague-Dawley , Receptor, Notch3/genetics , Signal Transduction , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Molecules ; 24(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627430

ABSTRACT

Twenty new 12N-substituted matrinol derivatives were synthesized and evaluated for their inhibitory effects on collagen α1 (I) (COL1A1) promotor in human hepatic stellate LX-2 cells. The structure-activity relationship (SAR) revealed that introducing a 12N-benzeneaminoacylmethyl substitution might significantly enhance the activity. Compound 8a exhibited the highest inhibitory potency against COL1A1, and its inhibition activity against COL1A1 was further confirmed on both the mRNA and protein levels. It also effectively inhibited the expression of α smooth muscle actin (α-SMA), fibronectin and transforming growth factor ß1 (TGFß1), indicating an extensive inhibitory effect on the expression of fibrogenic genes. The primary mechanism study indicated that it might take action via the Integrin/FAK/PI3K/Akt signaling pathway. The results provided powerful information for further structure optimization, and compound 8a was selected as a novel anti-fibrogenic lead for further investigation.


Subject(s)
Collagen Type I/genetics , Hepatic Stellate Cells/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Signal Transduction/drug effects , Thiazoles/pharmacology , Cell Line , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibrosis/prevention & control , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Humans , Integrin alphaV/genetics , Integrin alphaV/metabolism , Models, Biological , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Structure-Activity Relationship , Thiazoles/chemical synthesis
12.
Bioorg Chem ; 90: 103032, 2019 09.
Article in English | MEDLINE | ID: mdl-31207450

ABSTRACT

A series of new cytisine derivatives with a unique endocyclic scaffold were synthesized and evaluated for their inhibitory effect on collagen α1 (I) (COL1A1) promotor in human LX2 cells, taking cytisine as the lead. Structure-activity relationship (SAR) revealed that introducing a 12N-benzyl substitution might significantly enhance the activity. Compound 5f exhibited a promising inhibitory potency against COL1A1 with an IC50 value of 12.8 µM in human LX2 cells, and an inspiring inhibition activity against COL1A1 on both mRNA and protein levels. It also effectively inhibited the expression of α smooth muscle actin (α-SMA), connective tissue growth factor (CTGA), matrix metalloprotein 2 (MMP-2), and transforming growth factor ß1 (TGFß1), indicating an extensive inhibitory effect against fibrogenetic proteins. In addition, compound 5f displayed reasonable PK and safety profiles. The primary mechanism study indicated that it might repress the hepatic fibrogenesis via PI3K/Akt/Smad signaling pathway. The results provided powerful information for further structure optimization, and compound 5f was selected as a novel anti-liver fibrosis agent for further investigation.


Subject(s)
Alkaloids/therapeutic use , Liver Cirrhosis/drug therapy , Signal Transduction/drug effects , Alkaloids/chemical synthesis , Alkaloids/pharmacokinetics , Animals , Azocines/chemical synthesis , Azocines/pharmacokinetics , Azocines/therapeutic use , Cell Line , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Humans , Male , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolizines/chemical synthesis , Quinolizines/pharmacokinetics , Quinolizines/therapeutic use , Rats, Sprague-Dawley , Smad Proteins/metabolism , Structure-Activity Relationship
13.
Molecules ; 23(7)2018 07 05.
Article in English | MEDLINE | ID: mdl-29976890

ABSTRACT

A series of novel matrinic thiadiazole derivatives were designed, synthesized and evaluated for their inhibitory effect on COL1A1 promotor. The SAR indicated that: (i) the introduction of a thiadiazole on the 11-side chain was beneficial for activity; (ii) a 12-N-benzyl moiety was favorable for activity. Among them, compound 6n displayed a high activity with an inhibitory rate of 39.7% at a concentration of 40 µM. It also effectively inhibited the expression of two representative collagen proteins (COL1A1 and α-SMA) on both the mRNA and protein levels and showed a high safety profile in vivo, indicating its great promise as an anti-liver fibrosis agent. Further study indicated that it might repress hepatic fibrogenesis via the TGFß/Smad pathway. This study provided powerful information for further strategic optimization and the top compound 6n was selected for further study as an ideal liver fibrosis lead for next investigation.


Subject(s)
Actins/genetics , Alkaloids/chemistry , Collagen Type I/genetics , Quinolizines/chemistry , Thiadiazoles/administration & dosage , Thiadiazoles/chemical synthesis , Actins/pharmacology , Animals , Cell Line , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Hep G2 Cells , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Models, Biological , Molecular Structure , Signal Transduction/drug effects , Smad Proteins/metabolism , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Transforming Growth Factor beta/metabolism , Matrines
SELECTION OF CITATIONS
SEARCH DETAIL
...