Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Int J Mol Sci ; 24(21)2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37958659

ABSTRACT

Over the last four decades, vanadium compounds have been extensively studied as potential antidiabetic drugs. With the present review, we aim at presenting a general overview of the most promising compounds and the main results obtained with in vivo studies, reported from 1899-2023. The chemistry of vanadium is explored, discussing the importance of the structure and biochemistry of vanadate and the impact of its similarity with phosphate on the antidiabetic effect. The spectroscopic characterization of vanadium compounds is discussed, particularly magnetic resonance methodologies, emphasizing its relevance for understanding species activity, speciation, and interaction with biological membranes. Finally, the most relevant studies regarding the use of vanadium compounds to treat diabetes are summarized, considering both animal models and human clinical trials. An overview of the main hypotheses explaining the biological activity of these compounds is presented, particularly the most accepted pathway involving vanadium interaction with phosphatase and kinase enzymes involved in the insulin signaling cascade. From our point of view, the major discoveries regarding the pharmacological action of this family of compounds are not yet fully understood. Thus, we still believe that vanadium presents the potential to help in metabolic control and the clinical management of diabetes, either as an insulin-like drug or as an insulin adjuvant. We look forward to the next forty years of research in this field, aiming to discover a vanadium compound with the desired therapeutic properties.


Subject(s)
Diabetes Mellitus , Vanadium Compounds , Animals , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/chemistry , Vanadium Compounds/pharmacology , Vanadium Compounds/therapeutic use , Vanadium Compounds/chemistry , Vanadium/chemistry , Diabetes Mellitus/drug therapy , Insulin/therapeutic use , Insulin, Regular, Human/therapeutic use
2.
Int J Mol Sci ; 22(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34769364

ABSTRACT

Alzheimer's disease (AD) is an intractable neurodegenerative disease that leads to dementia, primarily in elderly people. The neurotoxicity of amyloid-beta (Aß) and tau protein has been demonstrated over the last two decades. In line with these findings, several etiological hypotheses of AD have been proposed, including the amyloid cascade hypothesis, the oxidative stress hypothesis, the inflammatory hypothesis, the cholinergic hypothesis, et al. In the meantime, great efforts had been made in developing effective drugs for AD. However, the clinical efficacy of the drugs that were approved by the US Food and Drug Association (FDA) to date were determined only mild/moderate. We recently adopted a vanadium compound bis(ethylmaltolato)-oxidovanadium (IV) (BEOV), which was originally used for curing diabetes mellitus (DM), to treat AD in a mouse model. It was shown that BEOV effectively reduced the Aß level, ameliorated the inflammation in brains of the AD mice, and improved the spatial learning and memory activities of the AD mice. These finding encouraged us to further examine the mechanisms underlying the therapeutic effects of BEOV in AD. In this review, we summarized the achievement of vanadium compounds in medical studies and investigated the prospect of BEOV in AD and DM treatment.


Subject(s)
Alzheimer Disease/drug therapy , Diabetes Mellitus/drug therapy , Vanadium Compounds/therapeutic use , Alzheimer Disease/pathology , Animals , Diabetes Mellitus/pathology , Humans
3.
Angew Chem Int Ed Engl ; 59(37): 15834-15838, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32598089

ABSTRACT

The chemistry and short lifetimes of metal-based anti-cancer drugs can be turned into an advantage for direct injections into tumors, which then allow the use of highly cytotoxic drugs. The release of their less toxic decomposition products into the blood will lead to decreased toxicity and can even have beneficial effects. We present a ternary VV complex, 1 ([VOL1 L2 ], where L1 is N-(salicylideneaminato)-N'-(2-hydroxyethyl)ethane-1,2-diamine and L2 is 3,5-di-tert-butylcatechol), which enters cells intact to induce high cytotoxicity in a range of human cancer cells, including T98g (glioma multiforme), while its decomposition products in cell culture medium were ≈8-fold less toxic. 1 was 12-fold more toxic than cisplatin in T98g cells and 6-fold more toxic in T98g cells than in a non-cancer human cell line, HFF-1. Its high toxicity in T98g cells was retained in the presence of physiological concentrations of the two main metal-binding serum proteins, albumin and transferrin. These properties favor further development of 1 for brain cancer treatment by intratumoral injections.


Subject(s)
Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Coordination Complexes/chemistry , Vanadium Compounds/chemistry , Antineoplastic Agents/therapeutic use , Brain Neoplasms/pathology , Cell Line, Tumor , Coordination Complexes/therapeutic use , Culture Media , Drug Discovery , Drug Screening Assays, Antitumor , Humans , Proton Magnetic Resonance Spectroscopy , Vanadium Compounds/therapeutic use
4.
Clin Sci (Lond) ; 130(5): 377-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26666444

ABSTRACT

Patients with diabetes are vulnerable to MI/R (myocardial ischaemia/reperfusion) injury, but are not responsive to IPostC (ischaemic post-conditioning) which activates PI3K (phosphoinositide 3-kinase)/Akt (also known as PKB or protein kinase B) and JAK2 (Janus kinase 2)/STAT3 (signal transducer and activator of transcription 3) pathways to confer cardioprotection. We hypothesized that increased cardiac PTEN (phosphatase and tensin homologue deleted on chromosome 10), a major negative regulator of PI3K/Akt, is responsible for the loss of diabetic heart sensitivity to IPostC cardioprotecton. In STZ (streptozotocin)-induced Type 1 diabetic rats subjected to MI/R (30 min coronary occlusion and 120 min reperfusion), the post-ischaemic myocardial infarct size, CK-MB (creatine kinase-MB) and 15-F2t-isoprostane release, as well as cardiac PTEN expression were significantly higher than those in non-diabetic controls, concomitant with more severe cardiac dysfunction and lower cardiac Akt, STAT3 and GSK-3ß (glycogen synthase kinase 3ß) phosphorylation. IPostC significantly attenuated post-ischaemic infarct size, decreased PTEN expression and further increased Akt, STAT3 and GSK-3ß phosphorylation in non-diabetic, but not in diabetic rats. Application of the PTEN inhibitor BpV (bisperoxovanadium) (1.0 mg/kg) restored IPostC cardioprotection in diabetic rats. HPostC (hypoxic post-conditioning) in combination with PTEN gene knockdown, but not HPostC alone, significantly reduced H/R (hypoxia/reoxygenation) injury in cardiac H9c2 cells exposed to high glucose as was evident from reduced apoptotic cell death and JC-1 monomer in cells, accompanied by increased phosphorylation of Akt, STAT3 and GSK-3ß. PTEN inhibition/gene knockdown mediated restoration of IPostC/HPostC cardioprotection was completely reversed by the PI3K inhibitor wortmannin, and partially reversed by the JAK2 inhibitor AG490. Increased cardiac PTEN, by impairing PI3K/Akt and JAK2/STAT3 pathways, is a major mechanism that rendered diabetic hearts not responsive to post-conditioning cardioprotection.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Ischemic Postconditioning/methods , Myocardial Reperfusion Injury/prevention & control , PTEN Phosphohydrolase/antagonists & inhibitors , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Knockdown Techniques/methods , Janus Kinase 2/physiology , Male , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Vanadium Compounds/pharmacology , Vanadium Compounds/therapeutic use
5.
Neurochem Res ; 40(8): 1593-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26081019

ABSTRACT

Early postnatal propofol administration has potential detrimental effects on hippocampal synaptic development and memory. Therapeutic method is still lack due to unknown mechanisms. In this study, a 7-day propofol protocol was applied to model anesthesia in neonatal mice. Phosphatase and tensin homolog deleted on chromosome ten (Pten) inhibitor bisperoxovanadium (bpV) was pre-applied before propofol to study its potential protection. After propofol application, Pten level increased while phospho-AKT (p-AKT) (Ser473) decreased in dorsal hippocampus. Interestingly, i.p. injection of Pten inhibitor reversed the decrease of p-AKT. Two months after administration, basal synaptic transmission, hippocampal long-term potentiation (LTP) and long-term memory were reduced in propofol-administrated mice. By contrast, i.p. injection of Pten inhibitor at a dose of 0.2 mg/kg/day before propofol reversed the detrimental effects due to propofol application. Consistently, bpV injection also reversed propofol application-induced decrease of synaptic plasticity-related proteins, including p-CamKIIα, p-PKA and postsynaptic density protein 95. Taken together, our results demonstrate that bpV injection could reverse early propofol exposure-induced decrease of memory and hippocampal LTP. bpV might be a potential therapeutic for memory impairment after early propofol postnatal application.


Subject(s)
Hippocampus/drug effects , Long-Term Potentiation/drug effects , Memory Disorders/prevention & control , PTEN Phosphohydrolase/antagonists & inhibitors , Propofol/toxicity , Vanadium Compounds/therapeutic use , Animals , Animals, Newborn , Female , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Memory Disorders/chemically induced , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Vanadium Compounds/pharmacology
6.
Int J Mol Sci ; 16(6): 12648-68, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26053397

ABSTRACT

This paper discusses how the activity and expression of cyclooxygenases are influenced by vanadium compounds at anticancer concentrations and recorded in inorganic vanadium poisonings. We refer mainly to the effects of vanadate (orthovanadate), vanadyl and pervanadate ions; the main focus is placed on their impact on intracellular signaling. We describe the exact mechanism of the effect of vanadium compounds on protein tyrosine phosphatases (PTP), epidermal growth factor receptor (EGFR), PLCγ, Src, mitogen-activated protein kinase (MAPK) cascades, transcription factor NF-κB, the effect on the proteolysis of COX-2 and the activity of cPLA2. For a better understanding of these processes, a lot of space is devoted to the transformation of vanadium compounds within the cell and the molecular influence on the direct targets of the discussed vanadium compounds.


Subject(s)
Inflammation Mediators/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Vanadium Compounds/pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/therapeutic use , Signal Transduction/drug effects , Vanadium Compounds/therapeutic use
7.
Heart Fail Rev ; 19(1): 123-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23430125

ABSTRACT

It is now well known that a cardiomyopathic state accompanies diabetes mellitus. Although insulin injections and conventional hypoglycemic drug therapy have been of invaluable help in reducing cardiac damage and dysfunction in diabetes, cardiac failure continues to be a common cause of death in the diabetic population. The use of alternative medicine to maintain health and treat a variety of diseases has achieved increasing popularity in recent years. The goal of alternative therapies in diabetic patients has been to lower circulating blood glucose levels and thereby treat diabetic complications. This paper will focus its discussion on the role of vanadium on diabetes and the associated cardiac dysfunction. Careful administration of a variety of forms of vanadium has produced impressive long-lasting control of blood glucose levels in both Type 1 and Type 2 diabetes in animals. This has been accompanied by, in many cases, a complete correction of the diabetic cardiomyopathy. The oral delivery of vanadium as a vanadate salt in the presence of tea has produced particularly impressive hypoglycemic effects and a restoration of cardiac function. This intriguing approach to the treatment of diabetes and its complications, however, deserves further intense investigation prior to its use as a conventional therapy for diabetic complications due to the unknown long-term effects of vanadium accumulation in the heart and other organs of the body.


Subject(s)
Complementary Therapies/methods , Diabetes Complications/therapy , Heart Diseases/therapy , Vanadium Compounds/therapeutic use , Heart Diseases/etiology , Humans , Treatment Outcome
8.
Int J Mol Sci ; 14(6): 12013-22, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739679

ABSTRACT

PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a dual-specificity lipid and protein phosphatase. The loss of PTEN was originally discovered in numerous human cancers. PTEN inhibition by bisperoxovanadium (bpV) reduces neurological damage after ischemic brain injury. The purpose of this study was to identify the optimal neuroprotective dose of bpV when administrated after focal ischemia/reperfusion (I/R) injury in rats. Focal I/R injury was induced using the middle cerebral artery occlusion method. bpV at doses of 0.25, 0.50 and 1.0 mg/kg were injected intraperitoneally just after reperfusion, with saline serving as a vehicle control. A maximal reduction in brain injury was observed with 1.0 mg/kg bpV. This dose of bpV also significantly blocked apoptosis in the penumbral cortex of rats. This beneficial effect was associated with the increasing levels of Akt phosphorylation in the penumbral cortex. These results demonstrate that the pharmacological inhibition of PTEN protects against I/R injury in a dose-dependent manner and the protective effect might be induced through upregulation of the phosphoinositide-3 kinase/Akt pro-survival pathway, suggesting a new therapeutic strategy to combat ischemic brain injury.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Vanadium Compounds/therapeutic use , Acute Disease , Animals , Apoptosis/drug effects , Blotting, Western , Brain Ischemia/pathology , Cerebral Infarction/complications , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , In Situ Nick-End Labeling , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Vanadium Compounds/pharmacology
9.
Cells ; 11(3)2022 02 07.
Article in English | MEDLINE | ID: mdl-35159385

ABSTRACT

Despite some significant advancements, breast cancer has become the most prevalent cancer in the world. One of the main reasons for failure in treatment and metastasis has been attributed to the presence of cancer initiating cells-cancer stem cells. Consequently, research is now being focussed on targeting cancer cells along with their stem cell population. Non-oncology drugs are gaining increasing attention for their potent anticancer activities. Metformin, a drug commonly used to treat type 2 diabetes, is the best example in this regard. It exerts its therapeutic action by activating 5' adenosine monophosphate-activated protein kinase (AMPK). Activated AMPK subsequently phosphorylates and targets several cellular pathways involved in cell growth and proliferation and the maintenance of stem-like properties of cancer stem cells. Therefore, AMPK is emerging as a target of choice for developing effective anticancer drugs. Vanadium compounds are well-known PTP inhibitors and AMPK activators. They find extensive applications in treatment of diabetes and obesity via PTP1B inhibition and AMPK-mediated inhibition of adipogenesis. However, their role in targeting cancer stem cells has not been explored yet. This review is an attempt to establish the applications of insulin mimetic vanadium compounds for the treatment of breast cancer by AMPK activation and PTP1B inhibition pathways.


Subject(s)
AMP-Activated Protein Kinases , Breast Neoplasms , Neoplastic Stem Cells , Vanadium Compounds , AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Neoplastic Stem Cells/metabolism , Vanadium Compounds/therapeutic use
10.
J Biol Inorg Chem ; 16(6): 961-72, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21667212

ABSTRACT

The interactions of metabolites of the antidiabetic vanadium-containing drug bis(maltolato)oxovanadium(IV) (BMOV) with lipid interface model systems were investigated and the results were used to describe a potentially novel mechanism by which these compounds initiate membrane-receptor-mediated signal transduction. Specifically, spectroscopic studies probed the BMOV oxidation and hydrolysis product interaction with interfaces created from cetyltrimethylammonium bromide (CTAB) which mimics the positively charged head group on phosphatidylcholine. (1)H and (51)V NMR spectroscopies were used to determine the location of the dioxobis(maltolato)oxovanadate(V) and the maltol ligand in micelles and reverse micelles by measuring changes in the chemical shift, signal linewidth, and species distribution. Both micelles and reverse micelles interacted similarly with the complex and the ligand, suggesting that interaction is strong as anticipated by Coulombic attraction between the positively charged lipid head group and the negatively charged complex and deprotonated ligand. The nature of the model system was confirmed using dynamic light scattering studies and conductivity measurements. Interactions of the complex/ligand above and below the critical micelle concentration of micelle formation were different, with much stronger interactions when CTAB was in the form of a micelle. Both the complex and the ligand penetrated the lipid interface and were located near the charged head group. These studies demonstrate that a lipid-like interface affects the stability of the complex and raise the possibility that ligand exchange at the interface may be important for the mode of action for these systems. Combined, these studies support recently reported in vivo observations of BMOV penetration into 3T3-L1 adipocyte membranes and increased translocation of a glucose transporter to the plasma membrane.


Subject(s)
Cell Membrane/chemistry , Coordination Complexes/chemistry , Hypoglycemic Agents/chemistry , Vanadium Compounds/chemistry , Caco-2 Cells , Cetrimonium , Cetrimonium Compounds/chemistry , Diabetes Mellitus/drug therapy , Humans , Hydrolysis , Hypoglycemic Agents/therapeutic use , Magnetic Resonance Spectroscopy , Micelles , Molecular Structure , Surface-Active Agents/chemistry , Vanadium Compounds/therapeutic use
11.
Mol Brain ; 14(1): 155, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34635126

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common motor neuron (MN) disease, with no present cure. The progressive loss of MNs is the hallmark of ALS. We have previously shown the therapeutic effects of the phosphatase and tensin homolog (PTEN) inhibitor, potassium bisperoxo (picolinato) vanadium (bpV[pic]), in models of neurological injury and demonstrated significant neuroprotective effects on MN survival. However, accumulating evidence suggests PTEN is detrimental for MN survival in ALS. Therefore, we hypothesized that treating the mutant superoxide dismutase 1 G93A (mSOD1G93A) mouse model of ALS during motor neuron degeneration and an in vitro model of mSOD1G93A motor neuron injury with bpV(pic) would prevent motor neuron loss. To test our hypothesis, we treated mSOD1G93A mice intraperitoneally daily with 400 µg/kg bpV(pic) from 70 to 90 days of age. Immunolabeled MNs and microglial reactivity were analyzed in lumbar spinal cord tissue, and bpV(pic) treatment significantly ameliorated ventral horn motor neuron loss in mSOD1G93A mice (p = 0.003) while not significantly altering microglial reactivity (p = 0.701). Treatment with bpV(pic) also significantly increased neuromuscular innervation (p = 0.018) but did not affect muscle atrophy. We also cultured motor neuron-like NSC-34 cells transfected with a plasmid to overexpress mutant SOD1G93A and starved them in serum-free medium for 24 h with and without bpV(pic) and downstream inhibitor of Akt signaling, LY294002. In vitro, bpV(pic) improved neuronal viability, and Akt inhibition reversed this protective effect (p < 0.05). In conclusion, our study indicates systemic bpV(pic) treatment could be a valuable neuroprotective therapy for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Motor Neurons/drug effects , Neuroprotective Agents/therapeutic use , Vanadium Compounds/therapeutic use , Amyotrophic Lateral Sclerosis/pathology , Animals , Anterior Horn Cells/drug effects , Cells, Cultured , Chromones/pharmacology , Culture Media, Serum-Free/pharmacology , Humans , Mice, Transgenic , Microglia/drug effects , Models, Animal , Morpholines/pharmacology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Mutation, Missense , Neuromuscular Junction/drug effects , Neuroprotective Agents/pharmacology , PTEN Phosphohydrolase/antagonists & inhibitors , Point Mutation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Superoxide Dismutase-1/deficiency , Superoxide Dismutase-1/genetics , Vanadium Compounds/pharmacology
12.
Am J Physiol Heart Circ Physiol ; 298(4): H1198-208, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097771

ABSTRACT

Phosphoinositide 3-kinase (PI3K) mediates myocardium protective signaling through phosphorylation of phosphatidylinositol (Ptdins) to produce Ptdins(3,4,5)P(3). Lipid phosphatase and tensin homolog on chromosome 10 (PTEN) antagonizes PI3K activity by dephosphorylating Ptdins(3,4,5)P(3); therefore, the inhibition of PTEN enhances PI3K/Akt signaling and could prevent myocardium from ischemia-reperfusion (I/R) injury. Here we studied 1) whether the pharmacological inhibition of PTEN by bisperoxovanadium molecules [BpV(HOpic)] attenuates simulated I/R (SIR) injury in vitro and 2) whether the administration of BpV(HOpic) either before or after ischemia limits myocardial infarct size (IS) and ameliorates cardiodysfunction caused by infarction. First, adult rat cardiomyocytes were treated with or without BpV(HOpic) and then exposure to SIR. Second, anesthetized rats received BpV(HOpic) either before or after ischemia. IS was assessed at 4 h reperfusion, and left ventricular function was evaluated by echocardiography at 28 days postreperfusion. As a result, BpV(HOpic) decreased cell death, improved 3-[4,5-yl]-2,5-diphenyltetrazolium bromide (MTT) viability, and reduced apoptosis in cells exposed to SIR. These protective effects of BpV(HOpic) are associated with increased phospho-Akt and the repression of caspase-3 activity. Second, the administration of BpV(HOpic) significantly reduced IS and suppressed caspase-3 activity following I/R injury and consequentially improved cardiac function at 28 day postinfarction. These beneficial effects of BpV(HOpic) are attributed to increases in myocardial levels of phosphorylation of Akt/endothelial nitric oxide synthase (eNOS), ERK-1/2, and calcium-dependent nitric oxide synthase activity. In conclusion, the pharmacological inhibition of PTEN protects against I/R injury through the upregulation of the PI3K/Akt/eNOS/ERK prosurvival pathway, suggesting a new therapeutic strategy to combat I/R injury.


Subject(s)
Enzyme Inhibitors/therapeutic use , Myocardial Infarction/drug therapy , PTEN Phosphohydrolase/antagonists & inhibitors , Vanadium Compounds/therapeutic use , Ventricular Dysfunction, Left/drug therapy , Animals , Caspase 3/metabolism , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type III/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/physiopathology , bcl-2-Associated X Protein/metabolism
13.
Biomed Mater ; 16(1): 014101, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33355313

ABSTRACT

In the past few decades, various vanadium compounds have displayed potential in cancer treatment. However, fast clearness in the body and possible toxicity of vanadium compounds has hindered their further development. Vanadium-based nanomaterials not only overcome these limitations, but take advantage of the internal properties of vanadium in photics and magnetics, which enable them as a multimodal platform for cancer diagnosis and treatment. In this paper, we first introduced the basic biological and pharmacological functions of vanadium compounds in treating cancer. Then, the synthesis routes of three vanadium-based nanomaterials were discussed, including vanadium oxides, 2D vanadium sulfides, carbides and nitrides: VmXn (X = S, C, N) and water-insoluble vanadium salts. Finally, we highlighted the applications of these vanadium-based nanomaterials as tumor therapeutic and diagnostic agents.


Subject(s)
Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Neoplasms/diagnosis , Neoplasms/therapy , Vanadium Compounds/chemistry , Vanadium Compounds/therapeutic use , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Contrast Media/chemistry , Humans , Luminescence , Magnetic Resonance Imaging/methods , Materials Testing , Metal Nanoparticles/ultrastructure , Microscopy, Electron, Transmission , Photothermal Therapy/methods , Vanadium Compounds/chemical synthesis
14.
J Inorg Biochem ; 208: 111094, 2020 07.
Article in English | MEDLINE | ID: mdl-32438270

ABSTRACT

Since the 1970s, the biological role of vanadium compounds has been discussed as insulin-mimetic or insulin-enhancer agents. The action of vanadium compounds has been investigated to determine how they influence the insulin signaling pathway. Khan and coworkers proposed key proteins for the insulin pathway study, introducing the concept "critical nodes". In this review, we also considered critical kinases and phosphatases that participate in this pathway, which will permit a better comprehension of a critical node, where vanadium can act: a) insulin receptor, insulin receptor substrates, and protein tyrosine phosphatases; b) phosphatidylinositol 3'-kinase, 3-phosphoinositide-dependent protein kinase and mammalian target of rapamycin complex, protein kinase B, and phosphatase and tensin homolog; and c) insulin receptor substrates and mitogen-activated protein kinases, each node having specific negative modulators. Additionally, leptin signaling was considered because together with insulin, it modulates glucose and lipid homeostasis. Even in recent literature, the possibility of vanadium acting against metabolic diseases or cancer is confirmed although the mechanisms of action are not well understood because these critical nodes have not been systematically investigated. Through this review, we establish that vanadium compounds mainly act as phosphatase inhibitors and hypothesize on their capacity to affect kinases, which are critical to other hormones that also act on common parts of the insulin pathway.


Subject(s)
Glucose/metabolism , Insulin/therapeutic use , Lipid Metabolism/drug effects , Metabolic Diseases , Neoplasms , Signal Transduction/drug effects , Vanadium Compounds/therapeutic use , Vanadium/therapeutic use , Animals , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
15.
Nanoscale ; 12(16): 9219-9230, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32307476

ABSTRACT

Vanadium compounds are promising anti-diabetic agents, and graphene quantum dots (GQDs) are emerging as potential drug delivery systems to improve drug solubility in water and membrane transport. Using highly dispersible and water-soluble GQDs, we herein prepared a novel GQD-VO (p-dmada) complex, in which vanadium coordination compounds [VO(p-dmada)] were packed closely on one side of the GQD sheets possibly via the π-π stacking mechanism. The in vitro tests showed that GQD-VO(p-dmada) exhibited membrane permeability (Papp) as good as that of GQDs with reduced cytotoxicity. In vivo tests on type 2 diabetic mice demonstrated that GQD-VO(p-dmada) exhibited a delayed glucose lowering profile but more profound effects on insulin enhancement and ß-cell protection after three-week treatment compared to VO(p-dmada) alone. In addition, GQD alone was observed for the first time to effectively lower the blood lipid levels of the db/db mice. Overall, GQD-VO(p-dmada) showed improved pharmacokinetic performance and hypoglycemic effects, and using GQD as a nanoplatform for drug delivery may provide vast opportunities for the further design of metal-based pharmaceutical agents.


Subject(s)
Graphite/chemistry , Hypoglycemic Agents/chemistry , Quantum Dots/chemistry , Vanadium Compounds/chemistry , Animals , Cell Membrane/metabolism , Cell Survival/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Dogs , Drug Delivery Systems , Graphite/pharmacokinetics , Graphite/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Lipids/blood , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic , Quantum Dots/therapeutic use , Solubility , Vanadium Compounds/pharmacokinetics , Vanadium Compounds/therapeutic use
16.
Biol Trace Elem Res ; 188(1): 68-98, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30350272

ABSTRACT

Vanadium compounds have been primarily investigated as potential therapeutic agents for the treatment of various major health issues, including cancer, atherosclerosis, and diabetes. The translation of vanadium-based compounds into clinical trials and ultimately into disease treatments remains hampered by the absence of a basic pharmacological and metabolic comprehension of such compounds. In this review, we examine the development of vanadium-containing compounds in biological systems regarding the role of the physiological environment, dosage, intracellular interactions, metabolic transformations, modulation of signaling pathways, toxicology, and transport and tissue distribution as well as therapeutic implications. From our point of view, the toxicological and pharmacological aspects in animal models and humans are not understood completely, and thus, we introduced them in a physiological environment and dosage context. Different transport proteins in blood plasma and mechanistic transport determinants are discussed. Furthermore, an overview of different vanadium species and the role of physiological factors (i.e., pH, redox conditions, concentration, and so on) are considered. Mechanistic specifications about different signaling pathways are discussed, particularly the phosphatases and kinases that are modulated dynamically by vanadium compounds because until now, the focus only has been on protein tyrosine phosphatase 1B as a vanadium target. Particular emphasis is laid on the therapeutic ability of vanadium-based compounds and their role for the treatment of diabetes mellitus, specifically on that of vanadate- and polioxovanadate-containing compounds. We aim at shedding light on the prevailing gaps between primary scientific data and information from animal models and human studies.


Subject(s)
Diabetes Mellitus/metabolism , Hypoglycemic Agents/therapeutic use , Vanadium Compounds/therapeutic use , Vanadium/chemistry , Vanadium/pharmacology , Animals , Diabetes Mellitus/drug therapy , Humans , Hypoglycemic Agents/chemistry , Vanadium/blood , Vanadium Compounds/chemistry
17.
Biomaterials ; 194: 94-104, 2019 02.
Article in English | MEDLINE | ID: mdl-30583152

ABSTRACT

Using metal oxide semiconductor nanomaterials for synergistic cancer treatment has recently attracted the attention of numerous researchers. Herein, oxygen-defective vanadium oxide nanodots (VOx NDs) with ultra-small size and great dispersibility were synthesized via a novel user-friendly method, and then doxorubicin was loaded onto the VOx NDs surfaces. The VOx NDs had great photothermal conversion efficiency and stability. Doxorubicin-loaded VOx NDs can simultaneously serve as therapeutic agent and tumor microenvironment-activable HSP60 inhibitor, resulting in improved efficacy of photothermal therapy and released active doxorubicin for chemotherapy. Finally, we show that synergistic treatment achieved significant therapeutic effects in mice. These results provided a promising strategy for developing novel methods of synthesizing metal oxide semiconductors for enhanced synergistic cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/therapy , Vanadium Compounds/therapeutic use , Animals , Chaperonin 60/antagonists & inhibitors , Chaperonin 60/metabolism , Green Chemistry Technology/methods , HCT116 Cells , Humans , Hyperthermia, Induced/methods , Mice , Mice, Nude , Nanotechnology/methods , Neoplasms/metabolism , Neoplasms/pathology , Oxides/therapeutic use
18.
Diabetes ; 56(2): 486-93, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259395

ABSTRACT

The hallmarks of insulin action are the stimulation and suppression of anabolic and catabolic responses, respectively. These responses are orchestrated by the insulin pathway and are initiated by the binding of insulin to the insulin receptor, which leads to activation of the receptor's intrinsic tyrosine kinase. Severe defects in the insulin pathway, such as in types A and B and advanced type 1 and 2 diabetes lead to severe insulin resistance, resulting in a partial or complete absence of response to exogenous insulin and other known classes of antidiabetes therapies. We have characterized a novel class of arylalkylamine vanadium salts that exert potent insulin-mimetic effects downstream of the insulin receptor in adipocytes. These compounds trigger insulin signaling, which is characterized by rapid activation of insulin receptor substrate-1, Akt, and glycogen synthase kinase-3 independent of insulin receptor phosphorylation. Administration of these compounds to animal models of diabetes lowered glycemia and normalized the plasma lipid profile. Arylalkylamine vanadium compounds also showed antidiabetic effects in severely diabetic rats with undetectable circulating insulin. These results demonstrate the feasibility of insulin-like regulation in the complete absence of insulin and downstream of the insulin receptor. This represents a novel therapeutic approach for diabetic patients with severe insulin resistance.


Subject(s)
Benzylamines/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Phosphoproteins/metabolism , Vanadium Compounds/therapeutic use , Adipocytes/drug effects , Administration, Oral , Animals , Benzylamines/chemistry , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Injections, Subcutaneous , Insulin Receptor Substrate Proteins , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Rats , Rats, Wistar , Streptozocin , Vanadium Compounds/chemistry
20.
Yakugaku Zasshi ; 128(3): 413-7, 2008 Mar.
Article in Japanese | MEDLINE | ID: mdl-18311061

ABSTRACT

Generation of neural precursors persists throughout life in the forebrain subventricular zone (SVZ) and dentate gyrus (DG) subgranular zone (SGZ) in rodent and human brains. In addition, newborn granule cells in the hippocampal DG are important for learning and memory formation. Brain injuries such as seizures or trauma could trigger endogenous programs for adult neurogenesis. Although brain ischemia also increases proliferation of neural progenitor cells in SVZ and SGZ, most neural progenitor cells are dead within 2 weeks after brain ischemia. In addition, there is no therapeutic agent to promote neurogenesis in the adult brain following brain injury. Here we found that intraperitoneal administrations of vanadium compounds, a stimulator of phosphatidylinositol 3-kinase (PI3K)/Akt and extracellular signal regulated kinase (ERK) pathways markedly enhances brain ischemia-induced neurogenesis. Thus, vanadium compounds are potential therapeutic agent to enhance ischemia-induced neurogenesis through PI3K/Akt and ERK activation.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Brain/physiology , Nerve Regeneration , Neurons/physiology , Vanadium Compounds/therapeutic use , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Injections, Intraperitoneal , Phosphatidylinositol 3-Kinases/metabolism , Stimulation, Chemical , Vanadium Compounds/administration & dosage , Vanadium Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL