Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 828
Filter
1.
Med Mycol ; 62(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39043448

ABSTRACT

Microsporidia are obligate, intracellular, spore-forming eukaryotic fungi that infect humans and animals. In the treatment of disseminated microsporidiosis albendazole is the choice of drug. In recent years, antiparasitic activity of phosphodiesterase (PDE) enzyme inhibitors has been demonstrated against parasites and fungi, however, there is no information on microsporidia. Vinpocetine is currently used as a cerebral vasodilator drug and also as a dietary supplement to improve cognitive functions. Vinpocetine inhibits PDE1, so we aimed to investigate whether vinpocetine alone or in combination with albendazole has any effect on the spore load of Encephalitozoon intestinalis (E. intestinalis)-infected HEK293 cells. After determining the noncytotoxic concentrations of vinpocetine and albendazole on the host cell by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, HEK293 cells were infected with E. intestinalis spores. Then, two different concentrations of vinpocetine, albendazole, and a combination of both drugs were applied to the cells with an interval of 72 h for 15 days. Spore load of the cells was analyzed by real-time PCR. After the last treatment, spore Deoxyribonucleic Acid (DNA) load was significantly reduced only in the group treated with 14 ng/ml albendazole. It was not different from control in groups treated with 7 ng/ml albendazole and 4-20 µM vinpocetine. However, the combination of vinpocetine significantly increased the effect of albendazole at both concentrations. To our knowledge, this is the first study to investigate the microsporicidal activity of vinpocetine as well as its combinations with albendazole. However, further studies are needed to investigate the mechanism of action and also confirm in vivo conditions.


Encephalitozoon intestinalis, a common cause of microsporidia-associated diseases in humans, albendazole is used in the treatment of E. intestinalis infection, vinpocetine inhibits PDE1 and voltage-gated Ca2+ channels, vinpocetine significantly enhances the effect of albendazole on E. intestinalis spore DNA load.


Subject(s)
Albendazole , Encephalitozoon , Vinca Alkaloids , Albendazole/pharmacology , Humans , Encephalitozoon/drug effects , Vinca Alkaloids/pharmacology , HEK293 Cells , Drug Synergism , Antifungal Agents/pharmacology , Spores, Fungal/drug effects , Cell Survival/drug effects
2.
Mol Biol Rep ; 51(1): 655, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38739285

ABSTRACT

BACKGROUND: There is limited data regarding the hazardous effect of gentamicin (GM) on the uterus and whether or not vinpocetine (Vinpo) ameliorates it. The present study aimed to identify the possible protective effect of Vinpo in GM-induced uterine injury in rats. METHODS: Female rats were assorted in control-group, Vinpo-group, GM-group, and Vinpo plus GM group. Serum and uterine GM concentration were measured. Uterine oxidative stress parameters besides inflammatory and apoptotic biomarkers were evaluated. Uterine histopathological examination and interlukin-1beta (IL-1ß) immune-histochemical study were detected. RESULTS: GM significantly increased uterine oxidative stress, inflammatory and apoptotic biomarkers. Histopathological picture of uterine damage and increased IL-1ß immunoexpression were detected. Vinpo significantly ameliorated the distributed GM concentration, oxidative stress, inflammatory and apoptotic biomarkers with a prompt improvement in histopathological picture and a decrease in IL-1ß immunoexpression. CONCLUSION: Vinpo protective effect against GM-induced uterine injury involves modulation of inflammasome/caspase-1/IL-1ß signaling pathway.


Subject(s)
Caspase 1 , Gentamicins , Inflammasomes , Interleukin-1beta , Oxidative Stress , Signal Transduction , Uterus , Vinca Alkaloids , Animals , Female , Interleukin-1beta/metabolism , Vinca Alkaloids/pharmacology , Rats , Caspase 1/metabolism , Gentamicins/adverse effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Uterus/drug effects , Uterus/metabolism , Uterus/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Apoptosis/drug effects
3.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619102

ABSTRACT

Tubulin-targeted chemotherapy has proven to be a successful and wide spectrum strategy against solid and liquid malignancies. Therefore, new ways to modulate this essential protein could lead to new antitumoral pharmacological approaches. Currently known tubulin agents bind to six distinct sites at α/ß-tubulin either promoting microtubule stabilization or depolymerization. We have discovered a seventh binding site at the tubulin intradimer interface where a novel microtubule-destabilizing cyclodepsipeptide, termed gatorbulin-1 (GB1), binds. GB1 has a unique chemotype produced by a marine cyanobacterium. We have elucidated this dual, chemical and mechanistic, novelty through multidimensional characterization, starting with bioactivity-guided natural product isolation and multinuclei NMR-based structure determination, revealing the modified pentapeptide with a functionally critical hydroxamate group; and validation by total synthesis. We have investigated the pharmacology using isogenic cancer cell screening, cellular profiling, and complementary phenotypic assays, and unveiled the underlying molecular mechanism by in vitro biochemical studies and high-resolution structural determination of the α/ß-tubulin-GB1 complex.


Subject(s)
Antineoplastic Agents/chemical synthesis , Bacterial Proteins/chemical synthesis , Biological Products/chemical synthesis , Depsipeptides/chemical synthesis , Microtubules/drug effects , Tubulin Modulators/chemical synthesis , Tubulin/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bacterial Proteins/isolation & purification , Bacterial Proteins/pharmacology , Binding Sites , Biological Products/isolation & purification , Biological Products/pharmacology , Cell Line, Tumor , Colchicine/chemistry , Colchicine/pharmacology , Crystallography, X-Ray , Cyanobacteria/chemistry , Depsipeptides/isolation & purification , Depsipeptides/pharmacology , Drug Discovery , HCT116 Cells , Humans , Maytansine/chemistry , Maytansine/pharmacology , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pyrones/chemistry , Pyrones/pharmacology , Taxoids/chemistry , Taxoids/pharmacology , Tubulin/genetics , Tubulin/metabolism , Tubulin Modulators/isolation & purification , Tubulin Modulators/pharmacology , Vinca Alkaloids/chemistry , Vinca Alkaloids/pharmacology
4.
Metab Brain Dis ; 38(6): 1831-1840, 2023 08.
Article in English | MEDLINE | ID: mdl-37335452

ABSTRACT

Vinpocetine (VPN) is an ethyl apovincaminate that has anti-inflammatory and antioxidant effects by inhibiting the expression of nuclear factor kappa B (NF-κB) and phosphodiesterase enzyme 1 (PDE-1). VPN is used in the management of stroke, dementia, and other neurodegenerative brain diseases. VPN may be effective in treating Parkinson's disease (PD). Therefore, this review aimed to clarify the mechanistic role of VPN in the management of PD. VPN has protective and restorative effects against neuronal injury by reducing neuroinflammation, and improvement of synaptic plasticity and cerebral blood flow. VPN protects dopaminergic neurons by reducing oxidative stress, lipid peroxidation, glutamate neurotoxicity, and regulation of Ca+ 2 overloads. VPN can alleviate PD neuropathology through its anti-inflammatory, antioxidant, antiapoptotic and neurogenic effects. VPN through inhibition of PDE1 improves cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP) signaling in the dopaminergic neurons of the substantia nigra (SN). VPN improves PD neuropathology through PDE1 inhibition with a subsequent increase of the cAMP/cGMP signaling pathway. Therefore, increasing cAMP leads to antioxidant effects, while augmentation of cGMP by VPN leads to anti-inflammatory effects which reduced neurotoxicity and development of motor severity in PD. In conclusion, this review indicated that VPN could be effective in the management of PD.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Vinca Alkaloids , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Antioxidants , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use , Dopaminergic Neurons
5.
Molecules ; 29(1)2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38202595

ABSTRACT

This study focuses on the synthesis of novel vinpocetine derivatives (2-25) and their biological evaluation. The chemical structures of the synthesized compounds were fully characterized using techniques such as 1H NMR, 13C NMR, and HRMS. The inhibitory activity of the synthesized compounds on PDE1A was evaluated, and the results revealed that compounds 3, 4, 5, 12, 14, 21, and 25 exhibited superior inhibitory activity compared to vinpocetine. Compound 4, with a para-methylphenyl substitution, showed a 5-fold improvement in inhibitory activity with an IC50 value of 3.53 ± 0.25 µM. Additionally, compound 25, with 3-chlorothiazole substitution, displayed an 8-fold increase in inhibitory activity compared to vinpocetine (IC50 = 2.08 ± 0.16 µM). Molecular docking studies were conducted to understand the binding models of compounds 4 and 25 within the active site of PDE1A. The molecular docking study revealed additional binding interactions, such as π-π stacking and hydrogen bonding, contributing to the enhanced inhibitory activity and stability of the ligand-protein complexes. Overall, the synthesized vinpocetine derivatives demonstrated promising inhibitory activity on PDE1A, and the molecular docking studies provided insights into their binding modes, supporting further development of these compounds as potential candidates for drug research and development.


Subject(s)
Indole Alkaloids , Vinca Alkaloids , Molecular Docking Simulation , Hydrogen Bonding , Vinca Alkaloids/pharmacology
6.
J Immunol ; 204(4): 933-942, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900337

ABSTRACT

Otitis media (OM) is the most common bacterial infection in children. It remains a major health problem and a substantial socioeconomic burden. Streptococcus pneumoniae (S. pneumoniae) is one of the most common bacterial pathogens causing OM. Innate inflammatory response plays a critical role in host defense against bacterial pathogens. However, if excessive, it has a detrimental impact on the middle ear, leading to middle ear inflammation, a hallmark of OM. Currently, there has been limited success in developing effective therapeutic agents to suppress inflammation without serious side effects. In this study, we show that vinpocetine, an antistroke drug, suppressed S. pneumoniae-induced inflammatory response in cultured middle ear epithelial cells as well as in the middle ear of mice. Interestingly, vinpocetine inhibited S. pneumoniae-induced inflammation via upregulating a key negative regulator cylindromatosis (CYLD). Moreover, CYLD suppressed S. pneumoniae-induced inflammation via inhibiting the activation of ERK. Importantly, the postinfection administration of vinpocetine markedly inhibited middle ear inflammation induced by S. pneumoniae in a well-established mouse OM model. These studies provide insights into the molecular mechanisms underlying the tight regulation of inflammation via inhibition of ERK by CYLD and identified vinpocetine as a potential therapeutic agent for suppressing the inflammatory response in the pathogenesis of OM via upregulating negative regulator CYLD expression.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Otitis Media/drug therapy , Pneumococcal Infections/drug therapy , Vinca Alkaloids/pharmacology , Animals , Cell Line , Deubiquitinating Enzyme CYLD/genetics , Disease Models, Animal , Ear, Middle/cytology , Ear, Middle/drug effects , Ear, Middle/immunology , Epithelial Cells , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Otitis Media/immunology , Otitis Media/microbiology , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , RNA, Small Interfering/metabolism , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/isolation & purification , Up-Regulation/drug effects , Vinca Alkaloids/therapeutic use
7.
Int J Mol Sci ; 23(18)2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36142133

ABSTRACT

Microtubule-targeting agents (MTAs) are effective drugs for cancer treatment. A novel diaryl [1,2]oxazole class of compounds binding the colchicine site was synthesized as cis-restricted-combretastatin-A-4-analogue and then chemically modified to have improved solubility and a wider therapeutic index as compared to vinca alkaloids and taxanes. On these bases, a new class of tricyclic compounds, containing the [1,2]oxazole ring and an isoindole moiety, has been synthetized, among which SIX2G emerged as improved MTA. Several findings highlighted the ability of some chemotherapeutics to induce immunogenic cell death (ICD), which is defined by the cell surface translocation of Calreticulin (CALR) via dissociation of the PP1/GADD34 complex. In this regard, we computationally predicted the ability of SIX2G to induce CALR exposure by interacting with the PP1 RVxF domain. We then assessed both the potential cytotoxic and immunogenic activity of SIX2G on in vitro models of multiple myeloma (MM), which is an incurable hematological malignancy characterized by an immunosuppressive milieu. We found that the treatment with SIX2G inhibited cell viability by inducing G2/M phase cell cycle arrest and apoptosis. Moreover, we observed the increase of hallmarks of ICD such as CALR exposure, ATP release and phospho-eIF2α protein level. Through co-culture experiments with immune cells, we demonstrated the increase of (i) CD86 maturation marker on dendritic cells, (ii) CD69 activation marker on cytotoxic T cells, and (iii) phagocytosis of tumor cells following treatment with SIX2G, confirming the onset of an immunogenic cascade. In conclusion, our findings provide a framework for further development of SIX2G as a new potential anti-MM agent.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Vinca Alkaloids , Humans , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Calreticulin/metabolism , Cell Line, Tumor , Colchicine/pharmacology , Immunogenic Cell Death , Isoindoles/pharmacology , Microtubules/metabolism , Multiple Myeloma/drug therapy , Oxazoles/pharmacology , Taxoids/pharmacology , Vinca Alkaloids/pharmacology , Pemetrexed/pharmacology , Pemetrexed/therapeutic use
8.
Int J Mol Sci ; 23(10)2022 May 18.
Article in English | MEDLINE | ID: mdl-35628467

ABSTRACT

Antimitotic agents such as the clinically approved vinca alkaloids, taxanes and epothilone can arrest cell growth during interphase and are therefore among the most important drugs available for treating cancer. These agents suppress microtubule dynamics and thus interfere with intracellular transport, inhibit cell proliferation and promote cell death. Because these drugs target biological processes that are essential to all cells, they face an additional challenge when compared to most other drug classes. General toxicity can limit the applicable dose and therefore reduce therapeutic benefits. Photopharmacology aims to avoid these side-effects by introducing compounds that can be applied globally to cells in their inactive form, then be selectively induced to bioactivity in targeted cells or tissue during a defined time window. This review discusses photoswitchable analogues of antimitotic agents that have been developed by combining different photoswitchable motifs with microtubule-stabilizing or microtubule-destabilizing agents.


Subject(s)
Antimitotic Agents , Antineoplastic Agents , Neoplasms , Vinca Alkaloids , Antimitotic Agents/metabolism , Antimitotic Agents/pharmacology , Antimitotic Agents/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Microtubules/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Vinca Alkaloids/metabolism , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use
9.
Inflammopharmacology ; 30(6): 2243-2259, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36190686

ABSTRACT

Traumatic brain damage is common worldwide and the treatments are not well-defined. Vinpocetine is a synthetic derivative of the vinca alkaloid vincamine and is clinically being used for various brain disorders. Here in the current study, we have investigated the neuroprotective potential of vinpocetine against traumatic brain injury. TBI was induced by the Marmarou weight drop method in rats. Brain damage was evaluated using cognitive and motor functions and the alterations in biomolecules. Injured rats were treated with different doses of vinpocetine (2.5, 5, and 10 mg/kg) for 4 weeks. Traumatic brain injury in rats produced significant deterioration of cognition and motor functions, which was accompanied by increased oxidative stress and significant alterations in brain monoamine levels as compared with the sham control group (p < 0.05). Vinpocetine alleviated TBI-induced oxidative burden, altered neurochemistry, and improved the cognitive and motor functions as compared with that of the TBI control group (p < 0.05). The observed neuroprotective potential of vinpocetine may be due to the observed antioxidant potential and its ability to restore the levels of brain neurochemicals under stressed conditions. The outcomes of the current study may help the repositioning of vinpocetine for preventing or treating traumatic brain injuries.


Subject(s)
Brain Injuries, Traumatic , Vinca Alkaloids , Animals , Rats , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use , Brain Injuries, Traumatic/drug therapy , Brain , Cognition
10.
Pharmazie ; 77(1): 9-13, 2022 01 03.
Article in English | MEDLINE | ID: mdl-35045919

ABSTRACT

This study aimed to examine the effects of vinpocetine on atopic dermatitis (AD) by administering it via oral, intraperitoneal, and topical routes to HR-1 hairless mice. AD was induced in the mice for five weeks with ovalbumin, and vinpocetine was administered twice daily through each route of administration for two weeks after the induction of AD. Vinpocetine (20, 10, and 2 mg/kg) was administered by oral, intraperitoneal, and topical routes, respectively. The administration of vinpocetine suppressed the increase in serum immunoglobulin (Ig) E and IgG1 levels and the production of interleukin (IL)-4 and IL-13-cytokines linked to T helper 2 cells in skin tissue. In addition, the invasion of inflammatory cells, including eosinophils, into the skin tissue was reduced, and changes in skin structure were also suppressed. These results show the potential for the use of vinpocetine in patients with AD and even for targeted treatment against PDE. In most of the experiments, symptom relief in the groups receiving oral and topical vinpocetine was slightly superior to that in the group receiving vinpocetine intraperitoneally. In particular, topical application of vinpocetine was found to be the most effective route when considering the dose of vinpocetine used in each route.


Subject(s)
Dermatitis, Atopic , Vinca Alkaloids , Animals , Cytokines , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Disease Models, Animal , Humans , Immunoglobulin E , Mice , Mice, Hairless , Skin , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use
11.
Mol Pharmacol ; 100(3): 181-192, 2021 09.
Article in English | MEDLINE | ID: mdl-34127539

ABSTRACT

Conophylline (CNP) is a vinca alkaloid extracted from the Tabernaemontana divaricata plant. It has been reported that CNP induces autophagy in a mammalian target of rapamycin-independent manner, and thereby inhibits protein aggregation. However, the mode of action of CNP in inducing autophagy remains unknown. In this study, we identified glutathione peroxidase 4 (GPX4) as a CNP-binding protein by using thermal proteome profiling. The technique exploits changes in the thermal stability of proteins resulting from ligand interaction, which is capable of identifying compound-binding proteins without chemical modification. GPX4, an antioxidant protein that uses reduced glutathione as a cofactor, directly catalyzes the reduction of hydrogen peroxide, organic hydroperoxides, and lipid peroxides. GPX4 suppresses lipid peroxide accumulation, and thus plays a key role in protecting cells from oxidative damage. We found that treatment with CNP caused accumulation of lipid reactive oxygen species (ROS) in cultured cells. Furthermore, similarly with CNP treatment, GPX4 deficiency caused accumulation of lipid ROS and induced autophagy. These findings indicate that GPX4 is a direct target of CNP involved in autophagy induction. SIGNIFICANCE STATEMENT: The present study identified glutathione peroxidase 4 (GPX4) as a binding protein of conophylline (CNP) by using thermal proteome profiling (TPP). This study showed that CNP treatment, similarly with the inhibition of GPX4, induced lipid reactive oxygen species accumulation and autophagy. The present findings suggest that GPX4 is the CNP target protein involved in autophagy induction. Furthermore, these results indicate that TPP is a useful technique for determining the mechanism of natural compounds.


Subject(s)
Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Proteomics/methods , Vinca Alkaloids/pharmacology , Autophagy/drug effects , Autophagy/physiology , Cell Line , Hot Temperature , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Anticancer Drugs ; 32(7): 727-733, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33735117

ABSTRACT

Vinpocetine is widely used to treat cerebrovascular diseases. However, the effect of vinpocetine to treat hepatocellular carcinoma (HCC) has not been investigated. In this study, we revealed that vinpocetine was associated with antiproliferative activity in HCC cells, but induced cytoprotective autophagy, which restricted its antitumor activity. Autophagy inhibitors improved the antiproliferative activity of vinpocetine in HCC cells. Sorafenib is effective to treat advanced HCC, but the effect of autophagy induced by sorafenib is indistinct. We demonstrated vinpocetine plus sorafenib suppressed the cytoprotective autophagy activated by vinpocetine in HCC cells and significantly induced apoptosis and suppressed cell proliferation in HCC cells. In addition, vinpocetine plus sorafenib activates glycogen synthase kinase 3ß (GSK-3ß) and subsequently inhibits cytoprotective autophagy induced by vinpocetine in HCC cells. Meanwhile, overexpression of GSK-3ß was efficient to increase the apoptosis induced by vinpocetine plus sorafenib in HCC cells. Our study revealed that vinpocetine plus sorafenib could suppress the cytoprotective autophagy induced by vinpocetine and subsequently show synergistically anti-HCC activity via activating GSK-3ß and the combination of vinpocetine and sorafenib might reverse sorafenib resistance via the PI3K/protein kinase B/GSK-3ß signaling axis. Thus, vinpocetine may be a potential candidate for sorafenib sensitization and HCC treatment, and our results may help to elucidate more effective therapeutic options for HCC patients with sorafenib resistance.


Subject(s)
Glycogen Synthase Kinase 3 beta/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Sorafenib/pharmacology , Vinca Alkaloids/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Proliferation/drug effects , Drug Therapy, Combination , Hep G2 Cells , Humans , Signal Transduction/drug effects , Sorafenib/administration & dosage , Vinca Alkaloids/administration & dosage
13.
Pharmacology ; 106(1-2): 37-44, 2021.
Article in English | MEDLINE | ID: mdl-32294652

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) originates in the nasopharyngeal epithelium. The most common treatments for NPC rT1-4 are radiotherapy and surgery. The pathogenesis of radiation-induced cognitive impairment is complex and includes oxidative stress, mitochondrial dysfunction, neuro-inflammation, and even apoptosis and cell death. Principally, toll-like receptors (TLRs) could regulate the inflammatory/anti-inflammatory balance in patients with radiation-induced brain injury. Vinpocetine has an anti-inflammatory effect as shown in both animal and in vitro studies. Also, dexamethasone is a widely used anti-inflammatory drug. Thus, it is important to test whether addition of vinpocetine could improve the anti-inflammatory properties of dexamethasone for the treatment of NPC patients with radiation-induced brain injuries. METHODS: A total of 60 NPC patients with radiation-related brain injury were recruited for this study. All subjects were randomly and blindly assigned to the following groups: the dexamethasone group (D group, n = 30) and the vinpocetine and dexamethasone group (VD group, n = 30). Both medicine treatments were uninterrupted for 14 days of administration. RESULTS: Combined administration of vinpocetine and dexamethasone lowered the expression levels of serum inflammatory cytokines, including TLR2, TLR4, interleukin (IL)-20, IL-8, tumor necrosis factor-α, interferon-γ, monocyte chemoattractant protein 2, and interferon-induced protein 20, when compared to dexamethasone monotherapy. Notably, combination therapy increased antioxidants (superoxide dismutase, glutathione, glutathione peroxidase, and glutathione reductase) and decreased oxidants (thiobarbituric acid reactive substances). Furthermore, combination therapy significantly increased the Mini Mental State Examination score, when compared to dexamethasone monotherapy. CONCLUSION: Administration of a combination of vinpocetine and dexamethasone may enhance the anti-inflammatory and anti-oxidative effects when compared to dexamethasone monotherapy, which leads to alleviated cognitive impairment in NPC patients with radiation injury.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cognitive Dysfunction/drug therapy , Dexamethasone/pharmacology , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/radiotherapy , Radiation Injuries/drug therapy , Vinca Alkaloids/pharmacology , Adult , Anti-Inflammatory Agents/therapeutic use , Antioxidants/metabolism , Cognition/drug effects , Cognitive Dysfunction/etiology , Cytokines/blood , Cytokines/drug effects , Dexamethasone/therapeutic use , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Inflammation/blood , Male , Mental Status and Dementia Tests , Middle Aged , Oxidants/metabolism , Oxidative Stress/drug effects , Radiation Injuries/complications , Random Allocation , Toll-Like Receptors/blood , Toll-Like Receptors/drug effects , Vinca Alkaloids/therapeutic use
14.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34302205

ABSTRACT

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Subject(s)
Angiotensin II/pharmacology , Antifibrotic Agents/pharmacology , Bone Morphogenetic Protein 4/genetics , Fibroblasts/drug effects , MAP Kinase Kinase 4/genetics , Vinca Alkaloids/pharmacology , Animals , Animals, Newborn , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Collagen/genetics , Collagen/metabolism , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Phosphorylation/drug effects , Primary Cell Culture , Rats , Rats, Wistar , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Clin Sci (Lond) ; 134(22): 2959-2976, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33111936

ABSTRACT

Abdominal aortic aneurysm (AAA), commonly occurring in the aged population, is a degenerative disease that dilate and weaken infrarenal aorta due to progressive degeneration of aortic wall integrity. Vinpocetine, a derivative of alkaloid vincamine, has long been used for cerebrovascular disorders and cognitive impairment in the aged population. Recent studies have indicated that vinpocetine antagonizes occlusive vascular disorders such as intimal hyperplasia and atherosclerosis. However, its role in vascular degenerative disease AAA remains unexplored. Herein, we determined the effect of vinpocetine on the formation of AAA as well as the intervention of pre-existing moderate AAA. AAA was induced by periaortic elastase application in C57BL/6J mice. Systemic vinpocetine treatment was applied daily via intraperitoneal injection. We showed that vinpocetine pre-treatment remarkably attenuated aneurysmal dilation assessed by diameter and volume. More importantly, vinpocetine also significantly suppressed the progression of pre-existing moderate AAA in a post-intervention model. Vinpocetine improved multiple cellular and molecular changes associated with AAA, such as elastin degradation, media smooth muscle cell depletion, collagen fibers remodeling and macrophage infiltration in aneurysmal tissues. Vinpocetine potently suppressed tumor necrosis factor-α-induced nuclear factor kappa-light-chain-enhancer of activated B cells activation and proinflammatory mediator expression in primary cultured macrophages in vitro, as well as in the aorta wall in vivo, suggesting vinpocetine conferred anti-AAA effect at least partially via the inhibition of inflammation. Taken together, our findings reveal a novel role of vinpocetine in AAA formation, development and progression. Given the excellent safety profile of vinpocetine, the present study suggests vinpocetine may be a novel therapeutic agent for AAA prevention and treatment.


Subject(s)
Aortic Aneurysm, Abdominal/drug therapy , Protective Agents/therapeutic use , Vinca Alkaloids/therapeutic use , Animals , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Dilatation, Pathologic , Disease Progression , Elastin/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Protective Agents/pharmacology , Proteoglycans/metabolism , Proteolysis/drug effects , Vinca Alkaloids/pharmacology
16.
J Cardiovasc Pharmacol ; 77(2): 208-216, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33351536

ABSTRACT

ABSTRACT: Ischemic stroke is the leading cause of globe death and permanent disability, but its therapeutic strategies are limited. Over the past decades, multiprotein complexes called inflammasomes have been shown as promising targets in ischemic stroke. Here, we examined vinpocetine (Vinp), a synthetic drug, playing a neuroprotective role against ischemic stroke in mice through regulating NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation. Middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to mimic ischemic stroke in vivo. Vinp was administrated by intraperitoneal injection with different dose (5 or 10 mg/kg) 1 hour after reperfusion. Then, neurological assessment and infarct size were performed, and interleukin-1ß (IL-1ß) and IL-18 levels were evaluated using ELISA. The levels of NLRP3 inflammasome components and its upstream nuclear factor-κB (NF-κB) were determined using real-time PCR or Western blot. The experimental results indicated that posttreatment with Vinp decreased cerebral infarct size, improved behavior recover, reduced NLRP3 inflammasome expression, and suppressed the transfer of NF-κB to nucleus and proinflammatory cytokine release in middle cerebral artery occlusion/reperfusion mice. In conclusion, this study demonstrates that Vinp alleviates ischemic stroke by regulating levels of NLRP3 inflammasome, NF-κB, and proinflammatory cytokines in vivo, offering an alternative medication for ischemic stroke associated with inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Infarction, Middle Cerebral Artery/prevention & control , Inflammasomes/metabolism , Ischemic Stroke/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotective Agents/pharmacology , Vinca Alkaloids/pharmacology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Inflammasomes/genetics , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction
17.
Bioorg Med Chem Lett ; 30(2): 126472, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31859156

ABSTRACT

A new series of Vinpocetine derivatives were synthesized and evaluated for their inhibitory activity on PDE1A in vitro. Seven compounds with higher inhibitory activity were selected for surface plasmon resonance (SPR) binding experiments. Compared with Vinpocetine, these high potency compounds presented a higher binding affinity with PDE1A, which was consistent with inhibitory activity. After further screening, compounds 5, 7, 21, 34 and Vinpocetine were selected to examine the vasorelaxant effects on endothelium-intact rat thoracic aortic rings. The study suggested that the effects of compounds 7 and 21 were the most significant with the maximum value of 93.46 ±â€¯0.77% and 92.90 ±â€¯0.78% (n = 5) at a concentration of 100 µM respectively. Based on these studies, compounds 7 and 21 were considered for further development as hit compounds.


Subject(s)
Vasodilator Agents/chemical synthesis , Vinca Alkaloids/chemistry , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Kinetics , Rats , Structure-Activity Relationship , Surface Plasmon Resonance , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology , Vinca Alkaloids/metabolism , Vinca Alkaloids/pharmacology
18.
J Biochem Mol Toxicol ; 34(10): e22555, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32578916

ABSTRACT

Hepatic damage is one of the most common complications related to cisplatin (Cis) use. Recently, liver protection lines are being discovered to avoid hepatic cell death as a result of oxidative, inflammatory, and apoptotic disturbance. Limited data reported the hepatoprotective effect of vinpocetine (Vin) in acute liver injury models. This study was designed to determine the potential protective effect of Vin (10-30 mg/kg, orally) against Cis-induced liver injury (10 mg/kg, IP) in mice. Vin administration for 1 week before Cis injection until the end of the experiment. On the 6th day after Cis injection, mice were anesthetized, blood and tissue samples were collected. Hepatic function, histological changes, oxidative stress, inflammation, and apoptotic markers were investigated. Vin administration ameliorated liver injury as indicated by decreased liver injury parameters; serum aminotransferases, ALK-P, GGT, and bilirubin, restored the anti-oxidant status by decrease MDA and NOx , and increased GSH and SOD, inhibited inflammation (IL-6, TNF-α, NFκB-p65, and iNOS) and apoptosis (Annexin-V, Bax, and Caspase-3) parameters. Vin confers dose-dependent protection against Cis-induced liver injury. The hepatoprotective effect of Vin involved anti-oxidative, anti-inflammatory, and anti-apoptotic activities.


Subject(s)
Annexin A5/metabolism , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Cisplatin/toxicity , Liver/drug effects , Vinca Alkaloids/pharmacology , bcl-2-Associated X Protein/metabolism , Animals , Biomarkers/blood , Body Weight/drug effects , Female , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Organ Size/drug effects
19.
J Biol Chem ; 293(52): 20214-20226, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30377255

ABSTRACT

Conophylline is a Vinca alkaloid from leaves of the tropical plant Ervatamia microphylla and has been shown to mimic the effect of the growth and differentiation factor activin A on pancreatic progenitor cells. However, activin A stimulates fibrosis of pancreatic stellate cells, whereas conophylline inhibits it, suggesting that this compound may serve as an antifibrotic drug. Here we investigated the effects of conophylline on human foreskin fibroblasts, especially focusing on extracellular matrix (ECM) proteins. A gene microarray analysis revealed that conophylline remarkably suppressed expression of the gene for hyaluronan synthase 2 (HAS2) and of its antisense RNA, whereas the expression of collagen genes was unaffected. Of note, immunostaining experiments revealed that conophylline substantially inhibits incorporation of versican and collagens into the ECM in cells treated with transforming growth factor ß (TGFß), which promotes collagen synthesis, but not in cells not treated with TGFß. Moreover, a protein biosynthesis assay disclosed that conophylline decreases collagen biosynthesis, concomitant with a decrease in total protein biosynthesis, indicating that conophylline-mediated inhibition of fibrosis is not specific to collagen synthesis. Conophylline affected neither TGFß-induced nuclear translocation of SMAD family member 2/3 (SMAD2/3) nor phosphorylation of SMAD2. However, conophylline substantially inhibited phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), suggesting that conophylline inhibits HAS2 expression via TGFß-mediated activation of the ERK1/2 pathway. Taken together, our results indicate that conophylline may be a useful inhibitor of ECM formation in fibrosis.


Subject(s)
Extracellular Matrix/metabolism , MAP Kinase Signaling System/drug effects , Vinca Alkaloids/pharmacology , Cells, Cultured , Collagen/metabolism , Fibroblasts , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hyaluronan Synthases/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Versicans/metabolism
20.
Epilepsia ; 60(12): 2459-2465, 2019 12.
Article in English | MEDLINE | ID: mdl-31755996

ABSTRACT

OBJECTIVE: To screen a library of potential therapeutic compounds for a woman with Lennox-Gastaut syndrome due to a Y302C GABRB3 (c.905A>G) mutation. METHODS: We compared the electrophysiological properties of cells with wild-type or the pathogenic GABRB3 mutation. RESULTS: Among 1320 compounds, multiple candidates enhanced GABRB3 channel conductance in cell models. Vinpocetine, an alkaloid derived from the periwinkle plant with anti-inflammatory properties and the ability to modulate sodium and channel channels, was the lead candidate based on efficacy and safety profile. Vinpocetine was administered as a dietary supplement over 6 months, reaching a dosage of 20 mg three times per day, and resulted in a sustained, dose-dependent reduction in spike-wave discharge frequency on electroencephalograms. Improved language and behavior were reported by family, and improvements in global impression of change surveys were observed by therapists blinded to intervention. SIGNIFICANCE: Vinpocetine has potential efficacy in treating patients with this mutation and possibly other GABRB3 mutations or other forms of epilepsy. Additional studies on pharmacokinetics, potential drug interactions, and safety are needed.


Subject(s)
Lennox Gastaut Syndrome/drug therapy , Lennox Gastaut Syndrome/genetics , Mutation/genetics , Precision Medicine/methods , Receptors, GABA-A/genetics , Vinca Alkaloids/therapeutic use , Adult , Dose-Response Relationship, Drug , Electroencephalography/drug effects , Electroencephalography/methods , Female , HEK293 Cells , Humans , Lennox Gastaut Syndrome/diagnosis , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Vinca Alkaloids/pharmacology , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL