Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 377: 114809, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714285

ABSTRACT

Neurogenesis as a potential strategy to improve the consequences of intracerebral hemorrhage (ICH). The current study investigates the effects of withaferin A (WFA) in combination with leptin (LEP) on ICH and neurogenesis mechanisms. LEP levels were dramatically reduced on days 7 and 14 following ICH insults in mice, but continuous WFA therapy significantly improved the potency of intrinsic LEP on day 14 after ICH. Furthermore, WFA combined with LEP enhances intrinsic neurogenesis and lessen motor deficits and long-term cognitive outcomes after ICH. In parallel, leptin deficiency in ob/ob mice limits enhancement of neurogenesis following ICH in response to WFA combined with LEP treatment. Importantly, the functional recovery conferred by WFA combined with LEP after ICH was inhibited by neurogenesis suppression. Mechanistically, this study unveiled that the signal transducer and activator of transcription-3 (STAT3) / suppressor of cytokine signaling-3 (SOCS3) pathway is a critical signaling pathway through which WFA combined with LEP treatment promotes intrinsic neurogenesis after ICH. Collectively, the results of this study elucidate the neuroprotective effects of WFA and LEP in ICH, and highlight a potential approach for ICH cell therapy.


Subject(s)
Cerebral Hemorrhage , Leptin , Mice, Inbred C57BL , Neurogenesis , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Withanolides , Animals , Withanolides/pharmacology , Neurogenesis/drug effects , STAT3 Transcription Factor/metabolism , Mice , Suppressor of Cytokine Signaling 3 Protein/metabolism , Leptin/pharmacology , Male , Signal Transduction/drug effects , Cerebral Hemorrhage/drug therapy , Neuroprotective Agents/pharmacology , Drug Therapy, Combination
2.
CNS Neurosci Ther ; 30(1): e14487, 2024 01.
Article in English | MEDLINE | ID: mdl-37803915

ABSTRACT

BACKGROUND: Chronic cerebral hypoperfusion-induced demyelination causes progressive white matter injury, although the pathogenic pathways are unknown. METHODS: The Single Cell Portal and PanglaoDB databases were used to analyze single-cell RNA sequencing experiments to determine the pattern of EAAT3 expression in CNS cells. Immunofluorescence (IF) was used to detect EAAT3 expression in oligodendrocytes and oligodendrocyte progenitor cells (OPCs). EAAT3 levels in mouse brains were measured using a western blot at various phases of development, as well as in traumatic brain injury (TBI) and intracerebral hemorrhage (ICH) mouse models. The mouse bilateral carotid artery stenosis (BCAS) model was used to create white matter injury. IF, Luxol Fast Blue staining, and electron microscopy were used to investigate the effect of remyelination. 5-Ethynyl-2-Deoxy Uridine staining, transwell chamber assays, and IF were used to examine the effects of OPCs' proliferation, migration, and differentiation in vivo and in vitro. The novel object recognition test, the Y-maze test, the rotarod test, and the grid walking test were used to examine the impact of behavioral modifications. RESULTS: A considerable amount of EAAT3 was expressed in OPCs and mature oligodendrocytes, according to single-cell RNA sequencing data. During multiple critical phases of mouse brain development, there were no substantial changes in EAAT3 levels in the hippocampus, cerebral cortex, or white matter. Furthermore, neither the TBI nor ICH models significantly affected the levels of EAAT3 in the aforementioned brain areas. The chronic white matter injury caused by BCAS, on the other hand, resulted in a strikingly high level of EAAT3 expression in the oligodendroglia and white matter. Correspondingly, blocking EAAT3 assisted in the recovery of cognitive and motor impairment as well as the restoration of cerebral blood flow following BCAS. Furthermore, EAAT3 suppression was connected to improved OPCs' survival and proliferation in vivo as well as faster OPCs' proliferation, migration, and differentiation in vitro. Furthermore, this study revealed that the mTOR pathway is implicated in EAAT3-mediated remyelination. CONCLUSIONS: Our findings provide the first evidence that abnormally high levels of oligodendroglial EAAT3 in chronic cerebral hypoperfusion impair OPCs' pro-remyelination actions, hence impeding white matter repair and functional recovery. EAAT3 inhibitors could be useful in the treatment of ischemia demyelination.


Subject(s)
Brain Injuries, Traumatic , Brain Ischemia , Carotid Stenosis , Demyelinating Diseases , Remyelination , White Matter , Animals , Mice , Brain Injuries, Traumatic/metabolism , Brain Ischemia/metabolism , Carotid Stenosis/pathology , Demyelinating Diseases/pathology , Mice, Inbred C57BL , Oligodendroglia/metabolism , White Matter/pathology
3.
Oncol Rep ; 49(4)2023 Apr.
Article in English | MEDLINE | ID: mdl-36866771

ABSTRACT

Subsequently to the publication of the above article, a concerned reader drew to our attention that the data panel shown in Fig. 7A for the 400 µM isoquercitrin experiment had previously appeared in Fig. 4A in another article published in the journal International Journal of Oncology [Tang B, Li Y, Yuan S, Tomlinson S and He S: Upregulation of the δ opioid receptor in liver cancer promotes liver cancer progression both in vitro and in vivo. Int J Oncol 43: 1281­1290, 2013], indicating that results that were purported to have been obtained under different experimental conditions had been derived from the same original source. Furthermore, concerns were also raised regarding the originality of some of the other data belonging to this figure. Given the errors that were identified in the compilation of Fig. 7 in this article, the Editor of Oncology Reports has decided that this article should be retracted from the publication owing to a lack of overall confidence in the presented data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience that might result from the retraction of this article. [Oncology Reports 31: 2377­2384, 2014; DOI: 10.3892/or.2014.3099].

4.
Cells ; 11(17)2022 09 03.
Article in English | MEDLINE | ID: mdl-36078162

ABSTRACT

Angiogenesis is a vital endogenous brain self-repair processes for neurological recovery after intracerebral hemorrhage (ICH). Increasing evidence suggests that leptin potentiates angiogenesis and plays a beneficial role in stroke. However, the proangiogenic effect of leptin on ICH has not been adequately explored. Moreover, leptin triggers post-ICH angiogenesis through pericyte, an important component of forming new blood vessels, which remains unclear. Here, we reported that exogenous leptin infusion dose-dependent promoted vascular endothelial cells survival and proliferation at chronic stage of ICH mice. Additionally, leptin robustly ameliorated pericytes loss, enhanced pericytes proliferation and migration in ICH mice in vivo, and in ICH human brain microvascular pericytes (HBVPC) in vitro. Notably, we showed that pericytes-derived pro-angiogenic factors were responsible for enhancing the survival, proliferation and tube formation followed leptin treatment in human brain microvascular endothelial cells (HCMEC/D3)/HBVPC co-culture models. Importantly, considerable improvements in neurobehavioral function and hostile microenvironment were observed in leptin treatment ICH mice, indicating that better vascular functionality post ICH improves outcome. Mechanistically, this study unveiled that leptin boost post-ICH angiogenesis potentially through modulation of leptin receptor (leptinR)/Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway in pericyte. Thus, leptin may be a lucrative option for the treatment of ICH.


Subject(s)
Cerebral Hemorrhage , Leptin , Neovascularization, Physiologic , Pericytes , Animals , Cerebral Hemorrhage/metabolism , Endothelial Cells/metabolism , Humans , Leptin/metabolism , Leptin/pharmacology , Mice , Pericytes/metabolism , STAT3 Transcription Factor/metabolism
5.
Mol Neurobiol ; 59(3): 1381-1397, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34993846

ABSTRACT

The pathogenic processes of brain injury after intracerebral hemorrhage (ICH) have not yet been fully elucidated. Increasing evidence suggests that ferroptosis activation aggravates injury after ICH, but the underlying mechanism remains unclear. Sphingosine kinase 1 (Sphk1) is a key enzyme in the regulation of sphingosine metabolism involved in the ferroptosis pathway, but its role in ICH needs clarification. In this study, transcriptional changes in ICH patients were assessed by microarray data, exposing Sphk1 as a highly upregulated gene during ICH. Furthermore, Sphk1 chemical inhibitors and siRNA were used to inhibit ICH-induced Sphk1 upregulation in in vivo and in vitro models, showing that Sphk1 inhibition after protects against ferroptosis and attenuates secondary brain injury and cell death. Mechanistically, this study unveiled that sphingosine kinase 1/sphingosine 1-phosphate/extracellular-regulated protein kinases/phosphorylated extracellular-regulated protein kinases (Sphk1/S1p/ERK/p-ERK) pathway is responsible for regulation of ferroptosis leading to secondary brain injury and cell death following ICH. Collectively, this study demonstrates that ferroptosis is closely associated with ICH, and that Sphk1 has a critical role in this lethal process. These results suggest a novel unique and effective therapeutic approach for ICH prevention and treatment.


Subject(s)
Brain Injuries , Ferroptosis , Brain Injuries/metabolism , Cerebral Hemorrhage/pathology , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/metabolism
6.
Molecules ; 26(19)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34641633

ABSTRACT

Due to the previous incorrect characterization of compound C1, the authors wish to make the following corrections to this paper [...].

7.
J Med Chem ; 64(9): 5485-5499, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33861929

ABSTRACT

Anticancer agents that present nonapoptotic cell death pathways are required for treating apoptosis-resistant pancreatic cancer. Here, we synthesized three fluorescent dithiocarbazate-copper complexes, {[CuII(L)(Cl)] 1, [CuII2(L)2(NO3)2] 2, and [CuII2CuI(L)2(Br)3] 3}, to assess their antipancreatic cancer activities. Complexes 1-3 showed significantly greater cytotoxicity toward several pancreatic cancer cell lines with better IC50 than those of the HL ligand and cisplatin. Confocal fluorescence imaging showed that complex 3 was primarily localized in the mitochondria. Primarily, compound 3 also can be applied to in vivo imaging. Further studies revealed that complex 3 kills pancreatic cancer cells by triggering multiple mechanisms, including ferroptosis. Complex 3 is the first copper complex to evoke cellular events consistent with ferroptosis in cancer cells. Finally, it significantly retarded the ASPC-1 cells' growth in a mouse xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Hydrazines/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Ferroptosis/drug effects , Half-Life , Humans , Hydrazines/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Mitochondria/chemistry , Mitochondria/metabolism , Molecular Conformation , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Transplantation, Heterologous
8.
Metallomics ; 11(11): 1847-1863, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31553341

ABSTRACT

To develop new anti-tumour Pt(ii) agents, we designed and synthesized five Pt(ii) complexes. These Pt(ii) complexes were modified benzene rings of 2-hydroxybenzylidene with a hydrocarbyl or halogen group. The five Pt(ii) complexes possessed remarkable cytotoxicity against tumour cells in vitro. In particular, we investigated chemotherapeutic mechanisms of the complexes against A549cisR cells. The Pt(ii) complexes could bind to and cleave DNA, while also inducing arrest of the cell cycle in S phase, leading to down-regulation of the levels of cyclin-dependent kinases and cyclin and up-regulation of the expression of p21. The selected complex, C3, changed the mitochondrial membrane potential and induced apoptosis. C3 also inhibited the expression of the c-myc gene and downstream proteins, thereby inhibiting telomerase activity.


Subject(s)
Hydrazones/chemical synthesis , Hydrazones/pharmacology , Platinum/pharmacology , Schiff Bases/chemical synthesis , Schiff Bases/pharmacology , Animals , Apoptosis/drug effects , Cattle , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Crystallography, X-Ray , DNA/metabolism , Humans , Inhibitory Concentration 50 , Ligands , Mitochondria/drug effects , Mitochondria/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Telomerase/antagonists & inhibitors , Telomerase/metabolism
9.
Molecules ; 24(14)2019 Jul 12.
Article in English | MEDLINE | ID: mdl-31336900

ABSTRACT

Multi-targeting of oncoproteins by a single molecule represents an effectual, rational, and an alternative approach to target therapy. We carried out a systematic study to reveal the mechanisms of action of newly synthesized Cu2+ compounds of 2-naphthalenol and 1-(((2-pyridinylmethyl)imino)methyl)- (C1 and C2). The antiproliferative activity of the as-synthesized complexes in three human cancer cell lines indicates their potential as multi-targeted antitumor agents. Relatively, C1 and C2 showed better efficacy in vitro relative to Cisplatin and presented promising levels of toxicity against A-549 cells. On the whole, the Cu2+ complexes exhibited chemotherapeutic effects by generating reactive oxygen species (ROS) and arresting the cell cycle in the G0/G1 phase by competent regulation of cyclin and cyclin-dependent kinases. Fascinatingly, the Cu2+ complexes were shown to activate the apoptotic and autophagic pathways in A-549 cells. These complexes effectively induced endoplasmic reticulum stress-mediated apoptosis, inhibited topoisomerase-1, and damaged cancer DNA through a ROS-mediated mechanism. The synthesized Cu2+ complexes established ROS-mediated targeting of multiple cell signaling pathways as a fabulous route for the inhibition of cancer cell growth.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Copper/chemistry , Naphthalenes/chemistry , Reactive Oxygen Species/metabolism , Autophagy/drug effects , Biomarkers , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Stability , Endoplasmic Reticulum Stress/drug effects , Humans , Molecular Structure , Signal Transduction/drug effects , Solubility , Structure-Activity Relationship
10.
Metallomics ; 11(8): 1372-1386, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31267119

ABSTRACT

Herein, to develop a multi-target anticancer metal agent and achieve a "1 + 1 > 2" pharmaceutical effect, we rationally designed and synthesized five complexes (C1-C5) by synergistically exploiting the properties of Zn(ii) and a series of modified 2,6-diacetylpyridine bis(thiosemicarbazone) ligands. By investigating the structure-activity relationships, we found that the binuclear Zn(ii) complex (C5) acts against human bladder cancer cells (T-24) with significant cytotoxicity. We subsequently determined the multiple anticancer mechanisms of C5 to T-24 cells, including inhibiting the activity of topoisomerase I (Topo I), blocking the cell cycle in the S phase, and inducing apoptosis and autophagy in T-24 cells. Furthermore, C5 inhibited the migration of T-24 cells and showed a significant cytostatic effect in the T-24 3D spheroid model.


Subject(s)
Antineoplastic Agents/pharmacology , Thiosemicarbazones/pharmacology , Urinary Bladder Neoplasms/drug therapy , Zinc/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , Humans , Models, Molecular , Thiosemicarbazones/chemistry , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Urinary Bladder Neoplasms/metabolism , Zinc/chemistry
11.
Eur J Med Chem ; 158: 442-452, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30241011

ABSTRACT

To develop potential next-generation metal anticancer agents, we designed and synthesised five Cu(II) 2-pyridine-thiosemicarbazone complexes by modifying the hydrogen atom at the N-4 position of ligands, and then investigated their structure-activity relationships and anticancer mechanisms. Modification of the N-4 position with different groups caused significant differences in cellular uptake and produced superior antitumor activity. Cu complexes arrested the cell cycle at S phase, leading to down-regulation of levels of cyclin and cyclin-dependent kinases and up-regulation of expression of cyclin-dependent kinase inhibitors. Cu complexes exerted chemotherapeutic effects via activating p53 and inducing production of reactive oxygen species to regulate expression of the B-cell lymphoma-2 family of proteins, causing a change in the mitochondrial membrane potential and release of cytochrome c to form a dimer with apoptosis protease activating factor-1, resulting in activation of caspase-9/3 to induce apoptosis. In addition, Cu complexes inhibited telomerase by down-regulating the c-myc regulator gene and expression of the human telomerase reverse transcriptase.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Copper/chemistry , Copper/pharmacology , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Drug Design , Humans , Ligands , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Eur J Med Chem ; 156: 368-380, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30015073

ABSTRACT

The ligand quantity, ligand type, and coordination geometry have important influences on the anticancer activity of metal-based complexes. On the basis of the structures of previously reported 1:1 Cu(II)/ligand complexes ([Cu(L1)Cl]·2H2O 1a, [Cu(L2)Cl]·H2O 2a, and [Cu(L2)NO3]·H2O 3a), we subsequently designed, developed, and characterized a series of corresponding 1:1:1 Cu(II)/ligand/co-ligand complexes ([Cu(L1)(Py)Cl]·H2O 1b, [Cu(L2)(Py)Cl] 2b, and [Cu(L2)(Py)NO3] 3b), where L1 = (E)-N'-(2-hydroxybenzylidene)acetohydrazide, L2 = (E)-N'-(2- hydroxybenzylidene)benzohydrazide, and Py = pyridine. All six Cu(II) complexes were assessed for their in vitro anticancer properties against a panel of human cancer cells, including cisplatin-resistant A549cisR cell lines. Interestingly, we observed that the 1:1:1 Cu/ligand/co-ligand mixed-ligand Cu(II) complexes exhibited higher anticancer activity than the corresponding 1:1 Cu(II)/ligand complexes. In particular, the 1:1:1 Cu(II)/ligand/co-ligand complex 3b displayed the greatest toxicity toward several cancer cells with better IC50 (1.12-3.77 µM) than cisplatin. Further mechanistic explorations showed that the 3b complex induced DNA damage, thus resulting in mitochondria-mediated apoptotic cell death. Furthermore, the 3b complex displayed pronounced cytostatic effects in the MCF-7 3D spheroid model.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Copper/chemistry , Copper/pharmacology , Hydrazones/chemistry , Hydrazones/pharmacology , Neoplasms/drug therapy , Apoptosis/drug effects , Cisplatin/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Crystallography, X-Ray , DNA Damage/drug effects , DNA Topoisomerases, Type I/metabolism , Humans , Ligands , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology
13.
J Inorg Biochem ; 185: 10-16, 2018 08.
Article in English | MEDLINE | ID: mdl-29730232

ABSTRACT

We synthesized and validated five Schiff base Pt(II) complexes derived from 2-hydroxy-1-naphthaldehyde benzoyl hydrazone and its derivatives, which are modified at the benzohydrazide structures (L1-L5). The complexes were [Pt(L1)(DMSO)Cl] (C1), [Pt(L2)(DMSO)Cl] (C2), [Pt(L3)(DMSO)Cl] (C3), [Pt(L4)(DMSO)Cl] (C4), and [Pt(L5)(DMSO)Cl] (C5). Crystal structures showed that the Pt centers of all complexes were tetra-coordinated with other atoms. The structure-activity relationships and anticancer mechanisms of the complexes were explored. These five Pt(II) complexes were toxic at micromolar doses and exhibited cytotoxicity similar to or somewhat higher than that of cisplatin, with IC50 values ranging from 4.38 µM to 25.16 µM. The complexes exerted chemotherapeutic effects via inhibition of telomerase by targeting the c-myc promoter and down-regulating the expression of human telomerase reverse transcriptase, consequently triggering cell apoptosis. In addition, Pt(II) complexes also caused cell cycle arrest at S-phase, leading to the down-regulation of cdc25 A, cyclin A2, and CDK2 and up-regulation of p53, p27, and p21 proteins. Other complex-associated events were reactive oxygen species production, transformation of the mitochondrial membrane potential (Δψm), release of cytochrome c, regulation of Bcl-2 family protein expression, facilitated release of apoptotic active substances, and activation of caspases to induce apoptosis.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Platinum Compounds/chemistry , Platinum Compounds/pharmacology , Stomach Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Reactive Oxygen Species/metabolism , Schiff Bases/chemistry , Spectrophotometry, Infrared , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Structure-Activity Relationship , Telomerase/metabolism
14.
Eur J Med Chem ; 134: 34-42, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28395152

ABSTRACT

Two types of 2-pyridinecarboxaldehyde thiosemicarbazones Ga(III) complexes, which are 2:1 and 1:1 ligand/Ga(III) complexes, were synthesized and determined by X-ray single crystal diffraction. The antiproliferative activity of these Ga(III) complexes have been examined to illuminate the structure-activity relationships essential to form Ga(III) complexes with remarkable anticancer activity. In addition, Ga(III) complexes where the metal/ligand ratio was 1:1 (C4) had observably higher antiproliferative activity than 1:2 (C3). Ga(III) complexes caused a marked increase of caspase-3 and 9 activity in NCI-H460 cells compared to the metal free ligand. Caspase activation was somewhat mediated by the release of Cyt C from mitochondria after incubation with selected agents. Both types of Ga(III) complexes showed more effective in inhibition of the G1/S transition than the ligand alone.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Gallium/pharmacology , Pyridines/pharmacology , Thiosemicarbazones/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Gallium/chemistry , Humans , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Pyridines/chemistry , Structure-Activity Relationship , Thiosemicarbazones/chemistry
15.
J Fluoresc ; 26(6): 1987-1996, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27488686

ABSTRACT

In an effort to better understand the biological efficacy of the tridentate aroyl hydrazone Cu(II) complexes, the Cu(II) complex of di-2-pyridyl ketone isonicotinoyl hydrazone ligand (HL), {[Cu(L)(H2O)]·H2O·NO3}n (C1) was synthesized and characterized. Single crystal X-ray study reveals that complex C1 forms 1D zigzag chains in solid state. In water, the hydrolysis of the 1D zigzag chains was observed, and finally formation of monomeric species. In vitro studies revealed that complex C1 showed significantly more anticancer activity than the ligand alone. Investigation of the anticancer mechanisms of C1, confirmed that the Cu(II) complex exhibit a strong capacity to promote productions of reactive oxygen species (ROS), leading to caspase-dependent apoptotic cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coordination Complexes/pharmacology , Copper/chemistry , Hydrazones/chemistry , Neoplasms/pathology , Pyridines/chemistry , Antineoplastic Agents/chemical synthesis , Cell Cycle/drug effects , Coordination Complexes/chemical synthesis , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Serum Albumin, Human/metabolism , Tumor Cells, Cultured
16.
Bioorg Med Chem ; 24(10): 2190-8, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27056799

ABSTRACT

In an effort to better understand the biological efficacy of the tridentate aroyl hydrazone Cu(II) complexes, three Cu(II) complexes of acetylpyridine benzoyl hydrazone (HL), [Cu(L)(NO3) (H2O)]·H2O (C1), [Cu(L)2] (C2) and [Cu(L)(HL)]·(NO3)(Sas) (C3) (Sas=salicylic acid) were synthesized and characterized. X-ray crystal structures and infrared (IR) spectra of the complexes reveal that the L(-) ligand of C1 and C2 are predominantly in the enolate resonance form, while one L(-) ligand in C3 is represented enolate resonance form and the other HL ligand exhibits keto resonance form. All Cu(II) complexes showed significantly more anticancer activity than the ligand alone. Interestingly, the Cu complexes where the ligand/metal ratio was 1:1 (C1) rather than 2:1 (C2 and C3) had higher antitumor efficacy. Moreover, the 1:1 Cu/ligand complex, C1, promotes A549 cell apoptosis possibly through the intrinsic reactive oxygen species (ROS) mediated mitochondrial pathway, accompanied by the regulation of Bcl-2 family proteins.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Copper/chemistry , Copper/pharmacology , Hydrazones/chemistry , Hydrazones/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/analogs & derivatives , Coordination Complexes/pharmacology , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
17.
Oncol Rep ; 31(5): 2377-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24676882

ABSTRACT

Liver cancer is a malignant tumour with high morbidity and fatality rates that is common worldwide. At present, the clinical approaches to treating primary liver cancer include partial hepatectomy, systemic or local chemotherapy, radiotherapy, radiofrequency ablative surgery and liver transplantation. However, all of these approaches have shortcomings, including poor prognosis and numerous side-effects. A large number of studies have proven that many effective ingredients in traditional Chinese medicine, particularly the flavonoid compounds extracted from plants, have achieved breakthroughs in terms of enhancing the effects and reducing the toxicity of chemotherapy and radiotherapy, preventing tumour metastasis and relapse after surgery, alleviating the clinical symptoms of advanced tumours, improving the quality of life of the patient with tumours and extending patient long­term survival. The purpose of the present study was to investigate the impact of isoquercitrin, the flavonoid from Bidens bipinnata L. extract, on the progression of liver cancer and to achieve a deeper understanding of the biological characteristics of isoquercitrin's involvement in the progression of liver cancer. In the in vitro experiments, isoquercitrin was found to strongly inhibit the proliferation of human liver cancer cells, promote the apoptosis of human liver cancer cells, and block the cell cycle in the G1 phase. Isoquercitrin activated caspase-3, -8 and -9, inhibited the expression level of ERK and p38MAPK protein phosphorylation, and promoted the phosphorylation of JNK. Additionally, isoquercitrin reduced the expression level of PKC in human liver cancer cells. In the in vivo experiments, isoquercitrin was also found to significantly inhibit the growth of transplanted tumours in nude mice. The present study confirmed that isoquercitrin could inhibit the progression of human liver cancer in vivo and in vitro, and the molecular mechanism of isoquercitrin may be closely associated with the MAPK and PKC signalling pathways.


Subject(s)
Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Quercetin/analogs & derivatives , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Extracellular Signal-Regulated MAP Kinases/biosynthesis , G1 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/prevention & control , Male , Medicine, Chinese Traditional , Mice , Mice, Nude , Phosphorylation , Quercetin/pharmacology , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Huazhong Univ Sci Technolog Med Sci ; 29(4): 431-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19662357

ABSTRACT

The current study was designed to determine the safety, tolerability and pharmacokinetic parameters of recombinant human parathyroid hormone [rhPTH (1-84)] used for the treatment of osteoporosis. In the single-dose format pharmacokinetic study, thirty-six healthy male volunteers received three dose levels of rhPTH (1-84) subcutaneously: 1, 2, and 4 microg/kg. The blood was timing drawn and the serum concentration of rhPTH (1-84) was determined by enzyme linked immunosorbent assay (ELISA). Serum concentration-time curves of PTH (1-84) exhibited a double-peak pattern, the first peak appearing about 10 to 30 min after administration and the second peak occurring about 1.5 to 2 h after administration. Serum terminal half-time of PTH (1-84) was approximately 2 h. The parameters indicated the serum levels were directly proportional to the administered dose, with the mean C(max) and AUC(0-24) ranging from approximately 543.47 to 1845 pg/mL and 2358.6 to 9232.12 pg.h.mL(-1) over the dose range. The drug was well tolerated, the clinical symptoms were generally mild and of short duration.


Subject(s)
Parathyroid Hormone/pharmacokinetics , Adult , China , Humans , Male , Parathyroid Hormone/adverse effects , Parathyroid Hormone/blood , Recombinant Proteins/adverse effects , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Young Adult
19.
Article in English | MEDLINE | ID: mdl-19224155

ABSTRACT

Batifiban, a synthetic cyclic peptide, is a potent platelet glycoprotein GPIIb/IIIa antagonist which may be useful in the treatment and prevention of acute coronary syndromes. The pharmacokinetics and pharmacodymanic (inhibition of platelet aggregation) effects, and tolerability of batifiban were investigated in healthy subjects following single bolus injection with doses of 55, 110, or 220 microg/kg, or multiple doses of an bolus followed intravenous infusion for 24 h (180 microg/kg plus 2.0 microg/min.kg, and 220 microg/kg plus 2.5 microg/min.kg) in this phase I clinical trial. Plasma levels of batifiban and areas under the curve were found to be proportional to doses. Batifiban was rapidly eliminated with a half-life of approximately 2.5 h. Significant differences were noted for plasma levels of batifiban and areas under the curve between males and females. No significant differences in the terminal half-life were found between males and females. Batifiban reversibly inhibited ex vivo platelet aggregation in a dose- and concentration-dependent manner, consistent with its mechanism as a GPIIb/IIIa antagonist. Single and multiple intravenous doses of batifiban were found to be safe and well tolerated in healthy subjects. These results support a bolus injection plus intravenous infusion regimen of batifiban for the treatment and prevention of acute coronary syndromes.


Subject(s)
Peptides, Cyclic/pharmacology , Peptides, Cyclic/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Adult , Female , Humans , Injections, Intravenous , Male , Platelet Aggregation Inhibitors/adverse effects , Young Adult
20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-301298

ABSTRACT

h. The parameters indicated the serum levels were directly proportional to the administered dose, with the mean Cmax and AUC0-24 ranging from approximately 543.47 to 1845 pg/mL and 2358.6 to 9232.12 mild and of short duration.

SELECTION OF CITATIONS
SEARCH DETAIL