Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Vasc Res ; 61(3): 109-121, 2024.
Article in English | MEDLINE | ID: mdl-38615660

ABSTRACT

INTRODUCTION: Following our recent finding that Ucp2 knockout promotes ferroptosis, we aimed to examine whether UCP2 alleviates myocardial ischemia/reperfusion injury (MI/RI) by inhibiting ferroptosis. METHODS: The left anterior descending coronary arteries of wild-type and Ucp2-/- C57BL/6 mice were ligated for 30 min and reperfused for 2 h to establish an MI/RI model. The effects of UCP2 on ferroptosis and MI/RI were determined by echocardiography, 2,3,5-triphenylttrazolium chloride staining, hematoxylin-eosin staining, Masson's trichrome staining, Sirius red staining, and analysis of myocardial injury markers and ferroptosis indicators. Ferrostatin-1 (Fer-1) and erastin (Era) were used to investigate whether UCP2 alleviated MI/RI by inhibiting ferroptosis and the molecular mechanism. RESULTS: UCP2 was upregulated in the MI/RI model in WT mice. Deletion of Ucp2 exacerbated ferroptosis, altered the expression levels of multiple ferroptosis-related genes, and significantly exacerbated MI/RI. Knockout of Ucp2 promoted ferroptosis induced by Era and inhibited the antiferroptotic effects of Fer-1. Knockout of Ucp2 activated the p53/TfR1 pathway to exacerbate ferroptosis. CONCLUSION: Our results showed that UCP2 inhibited ferroptosis in MI/RI, which might be related to regulation of the p53/TfR1 pathway.


Subject(s)
Disease Models, Animal , Ferroptosis , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury , Myocytes, Cardiac , Uncoupling Protein 2 , Animals , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/deficiency , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice
2.
Clin Exp Hypertens ; 46(1): 2326022, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38507311

ABSTRACT

BACKGROUND: Emodin is a traditional medicine that has been shown to exert anti-inflammatory and anti-oxidative effects. Previous research has indicated that emodin can alleviate myocardial remodeling and inhibit myocardial hypertrophy and fibrosis. However, the mechanism by which emodin affects myocardial fibrosis (MF) has not yet been elucidated. METHODS: Fibroblasts were treated with ANGII, and a mouse model of MF was established by ligation of the left anterior descending coronary artery. Cell proliferation was examined by a Cell Counting Kit-8 (CCK8) assay. Dihydroethidium (DHE) was used to measure reactive oxygen species (ROS) levels, and Masson and Sirius red staining were used to examine changes in collagen fiber levels. PI3K was over-expressed by lentiviral transfection to verify the effect of emodin on the PI3K/AKT/mTOR signaling axis. Changes in cardiac function in each group were examined by echocardiography. RESULTS: Emodin significantly inhibited fibroblast proliferation, decreased intracellular ROS levels, significantly upregulated collagen II expression, downregulated α-SMA expression, and inhibited PI3K/AKT/mTOR pathway activation in vitro. Moreover, the in vivo results were consistent with the in vitro. Emodin significantly decreased ROS levels in heart tissue and reduced collagen fibrillogenesis. Emodin could regulate the activity of PI3K to increase the expression of collagen II and downregulate α-SMA expression in part through the PI3K/AKT/mTOR pathway, and emodin significantly improved cardiac structure and function in mice. CONCLUSIONS: This study revealed that emodin targeted the PI3K/AKT/mTOR pathway to inhibit the development of myocardial fibrosis and may be an antifibrotic agent for the treatment of cardiac fibrosis.


Subject(s)
Emodin , Proto-Oncogene Proteins c-akt , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Emodin/pharmacology , Reactive Oxygen Species , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Fibrosis , Collagen
3.
J Fluoresc ; 28(4): 905-911, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29936595

ABSTRACT

A colorimetric and fluorescent indicator based on cinnamamide group-containing rhodamine derivative was synthesized for the detection of Hg2+. The rhodamine B and cinnamamide were connected via ethylenediamine as a bridging molecule through a condensation reaction to obtain a colorimetric and fluorescent indicator for the detection of Hg2+ in H2O-EtOH (4:1, v/v). The indicator was excellent in the selectivity of Hg2+ and was almost unaffected by other common ions such as Na+, K+, Mg2+, Fe3+, Cu2+, Zn2+, Cr3+. The Hg2+-containing aqueous solution turned from colorless to red within 7 min after the addition of the indicator, and had an absorption peak at 564 nm in UV-vis, which implies a significant colorimetric phenomenon. Their characteristic peaks varied with the Hg2+ content, and they reached a linear relationship at low concentrations. The binding stoichiometry proved to be 1:1. The lowest detection limit was 4.1 × 10-7 mol/L, ranging from acidic to neutral.

4.
J Fluoresc ; 28(3): 809-814, 2018 May.
Article in English | MEDLINE | ID: mdl-29808393

ABSTRACT

In this paper, a new kind of colorimetric chemsensor aiming at detecting Cr3+ has been synthesized, and it is based on the "Off-On" effect of a rhodamine derivative. Comparing with other metal irons (Na+, K+, Ni2+, Hg2+, Fe3+, Mn2+, Co2+, Cd2+, Cu2+, Pb2+, Zn2+, Mg2+, Ba2+, Ag+, Fe2+, Ce3+), the chemsensor has a quick and accurate response to Cr3+ in H2O-EtOH solution (4/1, v/v). There is an obvious change in color, from colorless to bright pink when Cr3+ is detected. According to the fitting curve based on Benesi-Hildebrand equation and working curve of absorption strength in UV-vis spectrum, the binding pattern of Cr3+ and the rhodamine derivative follows a 1:1 stoichiometry. The chemsensor shows great potential in monitoring Cr3+ in the aqueous medium with high efficiency, which is supposed to complete the recognition in the minimum as 5.2 × 10-7 mol/L within 5 min.

5.
Oncol Rep ; 45(6)2021 06.
Article in English | MEDLINE | ID: mdl-33846816

ABSTRACT

Colorectal cancer (CRC) is the third most diagnosed cancer worldwide. Progesterone is associated with a decreased risk of CRC and leads to a favourable prognosis. However, the specific mechanism by which progesterone suppresses malignant progression remains to be elucidated. In the present study, the level of progesterone was first analysed in 77 patients with CRC, and immunohistochemistry was performed to detect the expression of progesterone receptor (PGR) in the paired specimens. The correlations between progesterone, PGR and CRC prognosis were assessed. A Cell Counting Kit­8 assay was then used to detect proliferation of the CRC cells. Flow cytometry was performed to estimate apoptosis and to evaluate the cycle of the CRC cells. A xenograft tumour model was established in nude mice to assess the role of progesterone in tumour growth. Finally, a PCR microarray was used to screen differentially expressed genes to further interpret the mechanism by which progesterone inhibits the malignant progression of CRC. It was found that low expression of progesterone and PGR were significantly associated with poor prognosis of CRC. In addition, progesterone suppressed CRC cell proliferation by arresting the cell cycle and inducing apoptosis in vitro. Moreover, the inhibitory role of progesterone in tumour growth was verified in vivo. Further investigation showed that the level of growth arrest and DNA damage­inducible protein α (GADD45α) was up­regulated by progesterone, and this was followed by the activation of the JNK pathway. Progesterone increased the activity of the JNK pathway via GADD45α to inhibit proliferation by arresting the cell cycle and inducing apoptosis, thereby suppressing the malignant progression of CRC. Therefore, it can be concluded that progesterone and PGR might act as inhibiting factors for poor prognosis of CRC.


Subject(s)
Carcinoma/pathology , Colonic Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Progesterone/metabolism , Receptors, Progesterone/metabolism , Animals , Apoptosis/drug effects , Carcinoma/mortality , Carcinoma/surgery , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Colectomy , Colon/pathology , Colon/surgery , Colonic Neoplasms/mortality , Colonic Neoplasms/surgery , Female , Follow-Up Studies , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/surgery , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Progesterone/analysis , Prognosis , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Progesterone/analysis , Xenograft Model Antitumor Assays
6.
Biochem Pharmacol ; 177: 113946, 2020 07.
Article in English | MEDLINE | ID: mdl-32247852

ABSTRACT

Androgen receptor (AR) is a crucial driver of prostate cancer (PC). AR-relevant resistance remains a major challenge in castration-resistant prostate cancer (CRPC). Bromodomain and extra-terminal domain (BET) family are critical AR coregulators. Here, we developed several diphenylamine derivatives and identified compound 7d that disrupted the functions of AR and BET family in prostate cancer and exhibited favorable metabolic stability in vitro and high drug exposure in vivo. We showed 7d not only bound to AR, suppressed transactivation of wild-type AR (wt-AR) and the mutant that mediates Enzalutamide resistance, but also reduced c-Myc protein expression through BET inhibition. In addition, 7d inhibited the proliferation of AR-positive PC cells with favorable selectivity and suppressed AR-V7-expressing VCaP and 22Rv1 xenografts growth in vivo. Collectively, these results indicate the potential of lead compound 7d as an orally available AR and BET inhibitor to treat CRPC and overcome antiandrogen resistance.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Diphenylamine/pharmacology , Prostatic Neoplasms/drug therapy , Proteins/antagonists & inhibitors , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays/methods , Androgen Receptor Antagonists/chemical synthesis , Androgen Receptor Antagonists/chemistry , Animals , Cell Line, Tumor , Diphenylamine/chemical synthesis , Diphenylamine/chemistry , HEK293 Cells , HT29 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Models, Chemical , Molecular Structure , PC-3 Cells , Prostatic Neoplasms/metabolism , Proteins/metabolism
7.
J Med Chem ; 63(19): 11012-11033, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32924477

ABSTRACT

The nuclear protein poly(ADP-ribose) polymerase-1 (PARP1) has a well-established role in the signaling and repair of DNA and is a validated therapeutic target for cancers and other human diseases. Here, we have designed, synthesized, and evaluated a series of small-molecule PARP1 degraders based on the proteolysis-targeting chimera (PROTAC) concept. Our efforts have led to the discovery of highly potent PARP1 degraders, as exemplified by compound 18 (SK-575). SK-575 potently inhibits the growth of cancer cells bearing BRCA1/2 mutations and induces potent and specific degradation of PARP1 in various human cancer cells even at low picomolar concentrations. SK-575 achieves durable tumor growth inhibition in mice when used as a single agent or in combination with cytotoxic agents, such as temozolomide and cisplatin. These data demonstrate that SK-575 is a highly potent and efficacious PARP1 degrader.


Subject(s)
Antineoplastic Agents , Drug Design , Neoplasms , Phthalazines , Piperazines , Poly (ADP-Ribose) Polymerase-1 , Animals , Humans , Mice , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Ligands , Neoplasms/drug therapy , Phthalazines/chemistry , Piperazines/chemistry , Poly (ADP-Ribose) Polymerase-1/metabolism , Proteolysis
8.
Eur J Med Chem ; 187: 111952, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31846828

ABSTRACT

Cyclin-dependent kinase (CDK) family members are promising molecular targets in discovering potent inhibitors in disease settings, they function differentially. CDK2, CDK4 and CDK6, directly regulate the cell cycle, while CDK9 primarily modulates the transcription regulation. In discovering inhibitors of these CDKs, toxicity associated with off-target effect on other CDK homologs often posts as a clinical issue and hinders their further therapeutic development. To improve efficacy and reduce toxicity, here, using the Proteolysis Targeted Chimeras (PROTACs) approach, we design and further optimize small molecule degraders targeting multiple CDKs. We showed that heterobifunctional compound A9 selectively degraded CDK2. We also identified a dual-degrader, compound F3, which potently induced degradation of both CDK2 (DC50: 62 nM) and CDK9 (DC50: 33 nM). In human prostate cancer PC-3 cells, compound F3 potently inhibits cell proliferation by effectively blocking the cell cycle in S and G2/M phases. Our preliminary data suggests that PROTAC-oriented CDK2/9 degradation is potentially an effective therapeutic approach.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , MCF-7 Cells , Models, Molecular , Molecular Structure , PC-3 Cells , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Eur J Med Chem ; 171: 265-281, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30925341

ABSTRACT

Prostate cancer (PC) is the second most common malignancy in men worldwide. Among current therapies, two antiandrogens, Abiraterone Acetate and Enzalutamide (Enza) have become the standard of care for patients with metastatic castration-resistant prostate cancer (mCRPC). Here, we designed and synthesized a new series of nonsteroidal compounds deriving from the hybridization of Abiraterone (Abi) and Enzalutamide, among which compound 4a featuring the diphenylamine scaffold was identified as a potent and cell selective androgen receptor (AR) antagonist. In cell proliferation assays, compound 4a exhibited better antiproliferative activities than Enzalutamide against AR-overexpressing VCaP cells and 22Rv1 cells bearing H874Y-mutated AR. In addition, 4a suppressed the activity of AR-F876L mutant that confers resistance to Enzalutamide and efficiently blocked R1881-induced PSA and FKBP5 gene expression. In competitive binding assay, compound 4a displayed higher binding affinity to AR than Enzalutamide. These results suggest compound 4a as a potential candidate to treat Enza-resistant CRPC.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemical synthesis , Androgen Receptor Antagonists/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Models, Molecular , Molecular Structure , Prostatic Neoplasms, Castration-Resistant/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Eur J Med Chem ; 182: 111608, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31437779

ABSTRACT

Androgen receptor (AR) has been a target of prostate cancer (PC) for nearly six decades. Recently, downregulating or degrading AR and the mutants especially the splice variant 7 (AR-V7) lacking ligand binding domain (LBD) emerged as an advantageous therapeutic approach to overcome drug resistance. Here, the structural modification of darolutamide resulted in the discovery of dual-action AR inhibitors and down-regulators. Unlike other traditional AR antagonists targeting the AR-LBD, compounds 4k and 4b not only inhibit the activities of wt-AR and AR-F876L mutant but also downregulate the protein expression of full-length (AR-full) and AR variant 7 (AR-V7) at mRNA level. In cell proliferation assays, compounds 4k and 4b exhibited better antiproliferative activities than darolutamide and enzalutamide against AR-V7-positive 22Rv1 cells and VCaP cells. In addition, 4k demonstrated better antitumor activity than clinically used enzalutamide in castration-resistant VCaP xenograft model. Collectively, combining the activities of AR inhibition and downregulation, compound 4k is proposed as an advantageous lead compound to disrupt AR signaling and overcome resistance.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Drug Discovery , Pyrazoles/pharmacology , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemical synthesis , Androgen Receptor Antagonists/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Mutation , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Receptors, Androgen/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL