Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Wound Repair Regen ; 31(1): 77-86, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36484112

RESUMO

Wound dehiscence, oftentimes a result of the poor tensile strength of early healing wounds, is a significant threat to the post-operative patient, potentially causing life-threatening complications. Vanadate, a protein tyrosine phosphatase inhibitor, has been shown to alter the organisation of deposited collagen in healing wounds and significantly improve the tensile strength of incisional wounds in rats. In this study, we sought to explore the effects of locally administered vanadate on tensile strength and collagen organisation in both the early and remodelling phases of excisional wound healing in a murine model. Wild-type mice underwent stented excisional wounding on their dorsal skin and were divided equally into three treatment conditions: vanadate injection, saline injection control and an untreated control. Tensile strength testing, in vivo suction Cutometer analysis, gross wound measurements and histologic analysis were performed during healing, immediately upon wound closure, and after 4 weeks of remodelling. We found that vanadate treatment significantly increased the tensile strength of wounds and their stiffness relative to control wounds, both immediately upon healing and into the remodelling phase. Histologic analysis revealed that these biomechanical changes were likely the result of increased collagen deposition and an altered collagen organisation composed of thicker and distinctly organised collagen bundles. Given the risk that dehiscence poses to all operative patients, vanadate presents an interesting therapeutic avenue to improve the strength of post-operative wounds and unstable chronic wounds to reduce the risk of dehiscence.


Assuntos
Ferida Cirúrgica , Cicatrização , Ratos , Camundongos , Animais , Vanadatos/farmacologia , Vanadatos/metabolismo , Vanadatos/uso terapêutico , Modelos Animais de Doenças , Resistência à Tração , Colágeno/metabolismo , Pele/lesões , Ferida Cirúrgica/metabolismo
2.
Neurochem Res ; 46(5): 1151-1165, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33559829

RESUMO

The consumption of foods rich in carbohydrates, saturated fat, and sodium, accompanied by a sedentary routine, are factors that contribute to the progress of metabolic syndrome (MS). In this way, they cause the accumulation of body fat, hypertension, dyslipidemia, and hyperglycemia. Additionally, MS has been shown to cause oxidative stress, inflammation, and death of neurons in the hippocampus. Consequently, spatial and recognition memory is affected. It has recently been proposed that metformin decavanadate (MetfDeca) exerts insulin mimetic effects that enhance metabolism in MS animals; however, what effects it can cause on the hippocampal neurons of rats with MS are unknown. The objective of the work was to evaluate the effect of MetfDeca on hippocampal neurodegeneration and recognition memory in rats with MS. Administration of MetfDeca for 60 days in MS rats improved object recognition memory (NORt). In addition, MetfDeca reduced markers of oxidative stress and hippocampal neuroinflammation. Accompanied by an increase in the density and length of the dendritic spines of the hippocampus of rats with MS. We conclude that MetfDeca represents an important therapeutic agent to treat MS and induce neuronal and cognitive restoration mechanisms.


Assuntos
Memória/efeitos dos fármacos , Síndrome Metabólica/tratamento farmacológico , Metformina/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Vanadatos/uso terapêutico , Animais , Catalase/metabolismo , Combinação de Medicamentos , Hipocampo/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/patologia , Masculino , Síndrome Metabólica/complicações , Síndrome Metabólica/patologia , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , Superóxido Dismutase/efeitos dos fármacos
3.
Brain Inj ; 33(10): 1385-1401, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31319723

RESUMO

Primary Objective: Eph/Ephrin signaling is inhibitory for developing axons and blocking Eph pathways enhances regeneration after spinal cord injury. It was hypothesized that inhibition of Eph signaling promotes cellular and behavioral recovery after traumatic brain injury (TBI). Research design: Lateral fluid percussion (LFP) injury was performed on wildtype (WT) and EphA6 knockout (KO) mice. EphA6-Fc, Ephrin-A5-Fc fusion proteins, and sodium orthovanadate were used to alter the signaling pathway. Immunohistochemistry and tissue explants revealed cellular changes. Rotarod tests demonstrated vestibulomotor function. Outcomes: The EphA6 receptor expression is upregulated following LFP. Uninjured EphA6 KO mice exhibit greater neurite density and clustered Ephrin-A5-Fc causes growth cone collapse in vitro. After LFP, EphA6 KO mice demonstrate longer neurites and decreased neuronal cell death and astrocytosis compared to WT mice. Blocking EphA signaling by soluble EphA6-Fc fusion protein reduces cell death and improves motor function following LFP whereas clustered Ephrin-A5-Fc exacerbates cell death and neurodegeneration. Sodium orthovanadate rescues growth cone collapse in vitro as well as cell death and neurodegeneration in vivo. Conclusions: Eph/Ephrin signaling plays an inhibitory role following TBI. Targeting the Eph signaling pathway with Fc fusion proteins and pharmacological agents can be a novel strategy to counter the damaging effects of TBI. Abbreviations: LFP: lateral fluid percussion; TBI: traumatic brain injury; KO: knockout; WT: wildtype; PTP2: protein phosphotyrosine phosphatase 2; Tg: transgenic; YFP: yellow fluorescent protein; ATM: atmospheres; RT-qPCR: Real-time-quantitative PCR; dpi: days post injury; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; DAPI: 4',6-diamidino-2-phenylindole; PBS: phosphate buffered saline; GFAP: glial fibrillary acidic protein; FLJC: fluorojade C; CA: cornu ammonis; SEM: standard error of the mean; ANOVA: analysis of variance; PLSD: posthoc least significant difference.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/terapia , Terapia Genética/métodos , Receptor EphA1/antagonistas & inibidores , Receptor EphA1/genética , Animais , Astrócitos/patologia , Lesões Encefálicas Traumáticas/patologia , Morte Celular , Imunoglobulina G/farmacologia , Masculino , Camundongos , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/prevenção & controle , Neuritos/patologia , Neurônios/metabolismo , Equilíbrio Postural , Receptor EphA1/biossíntese , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Vanadatos/uso terapêutico
4.
Biochem Biophys Res Commun ; 501(3): 771-778, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29763604

RESUMO

Accumulation and polarization of anti-inflammatory M2 to proinflammatory M1 macrophage in the adipose tissue of obese diabetic mice is an important pathogenic signature. It worsens lipid induced inflammation and insulin resistance. Here we demonstrate that a small molecule, a peroxyvanadate compound i.e. DmpzH [VO(O2)2 (dmpz)] or dmp, could robustly decrease macrophage infiltration, accumulation and their polarization in high fat diet (HFD) induced obese diabetic mice. In searching the underlying mechanism it was revealed that SIRT1 level was strikingly low in the inflamed adipose tissue of HFD mice as compared to mice fed with standard diet (SD). Administration of dmp markedly increased SIRT1 level by inducing its gene expression with a consequent decrease in macrophage population. Elevation of SIRT1 coincided with the decrease of MCP1, Fetuin-A (FetA) and IFNγ. Since MCP1 and FetA drive macrophage to inflamed adipose tissue and IFNγ promotes M2 to M1 transformation, both recruitment and M1 induced inflammation were found to be significantly repressed by dmp. In addressing the question about how dmp induced excess SIRT1 could reduce MCP1, FetA and IFNγ levels, we found that it was due to the inactivation of NFκB because of its deacetylation by SIRT1. Since NFκB is the transcriptional regulator of these molecules, their expressions were significantly suppressed and that caused sharp decline in macrophage recruitment and their polarity to M1. This effected a marked fall in proinflammatory cytokine level which significantly improved insulin sensitivity. dmp is likely to be the first molecule that rescues inflammatory burden contributed by macrophage in obese diabetic mice adipose tissue which causes significant increase in insulin sensitivity therefore it may be a meaningful choice to treat type 2 diabetes.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Resistência à Insulina , Macrófagos/efeitos dos fármacos , Obesidade/complicações , Obesidade/tratamento farmacológico , Vanadatos/uso terapêutico , Animais , Polaridade Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Obesos , Obesidade/patologia , Células RAW 264.7
5.
J Surg Res ; 207: 102-107, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27979464

RESUMO

BACKGROUND: There are over two million laparotomies performed in the United States each year with an incisional hernia rate between 2% and 11%. A total of 100,000 ventral hernia repairs are undertaken each year with recurrences as high as 50%. MATERIALS AND METHODS: Full thickness midline fascia incisions from the xiphoid to the pubic symphysis were made in rats. The fascia and/or muscular layer was sutured closed and a gel with 300 µM of sodium orthovanadate or saline was placed over the suture line with the skin closed over it. On day 10, 1-cm strips from the superior, middle, and inferior regions of the abdominal wall were tested for breaking strength and processed for histology. RESULTS: The mean wound breaking strength of vanadate-treated wounds was 18.6 ± 2.7 N compared with 9.4 ± 3.6 N for controls (P < 0.0001). Similar quantities of granulation tissue were deposited in treated and control wounds. Fine green birefringence patterns, characteristic of immature connective tissue, were seen in control samples viewed with polarized light. In contrast, vanadate-treated wounds showed thick yellow-orange birefringence patterns characteristics of more mature connective tissue. Using α-smooth muscle actin immunostaining, myofibroblasts were prominent in control incisions, but few were identified in vanadate-treated incisions. CONCLUSIONS: In rat laparotomy wounds, a single application of vanadate increases wound breaking strength, through enhanced connective tissue organization. These combined data suggest topical application of vanadate immediately after fascial closure will increase wound strength, possibly reducing hernia recurrences in the repaired abdominal wall.


Assuntos
Técnicas de Fechamento de Ferimentos Abdominais , Inibidores Enzimáticos/uso terapêutico , Hérnia Incisional/prevenção & controle , Laparotomia , Ferida Cirúrgica/tratamento farmacológico , Vanadatos/uso terapêutico , Administração Tópica , Animais , Fenômenos Biomecânicos , Tecido Conjuntivo/efeitos dos fármacos , Tecido Conjuntivo/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Ferida Cirúrgica/fisiopatologia , Resistência à Tração/efeitos dos fármacos , Resultado do Tratamento , Vanadatos/farmacologia , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
6.
J Stroke Cerebrovasc Dis ; 24(11): 2434-46, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26324516

RESUMO

BACKGROUND: Estrogen deficiency and increase in protein tyrosine phosphatase (PTPase) activity may be a key mechanism in postmenopausal dyslipidemia-induced vascular dysfunction and dementia. Thus, the present study has been designed to investigate the effect of biochanin A (BCA, a phytoestrogen) and sodium orthovanadate (SOV), an inhibitor of PTPase in dyslipidemia-induced vascular dementia in ovariectomized rats. METHODS: Female Wistar rats were ovariectomized and fed on high fat diet for 4 weeks to produce dyslipidemia. Dyslipidemia was assessed by estimation of serum lipid levels including total cholesterol, triglyceride, HDL, and LDL levels. Dementia was assessed in terms of increase in brain acetylcholinesterase (AChE) activity and attenuation of learning ability (escape latency time) and memory retention (time spent in target quadrant) using Morris water maze. Vascular dysfunction was assessed in terms of attenuation of acetylcholine-induced endothelium-dependent relaxation (isolated carotid ring preparation), mRNA expression of endothelial nitric oxide synthase, and increase in serum thiobarbituric acid reactive species, superoxide anion level. Neurodegeneration was assessed in hippocampus by hematoxylin and eosin staining. BCA (2.5 and 5 mg/kg) and SOV (5 and 10 mg/kg) were administered alone and in low-dose combination to ovariectomized dyslipidemic rats. RESULTS: BCA (2.5 and 5 mg/kg), SOV (5 and 10 mg/kg), and donepezil (1 mg/kg) significantly improves vascular function, and learning and memory ability and decreases the neuronal cell death, oxidative stress, and AChE in ovariectomized dyslipidemic rats. CONCLUSIONS: Thus, it may be concluded that BCA and SOV attenuate vascular dysfunction and dementia in dyslipidemic ovariectomized rats.


Assuntos
Demência Vascular/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Genisteína/uso terapêutico , Fitoestrógenos/uso terapêutico , Vanadatos/uso terapêutico , Acetilcolinesterase/metabolismo , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Demência Vascular/etiologia , Donepezila , Dislipidemias/complicações , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Indanos/farmacologia , Indanos/uso terapêutico , Lipídeos/sangue , Rememoração Mental/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Ovariectomia , Estresse Oxidativo/efeitos dos fármacos , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Vanadatos/farmacologia
7.
Cancer Chemother Pharmacol ; 93(1): 55-70, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37755518

RESUMO

BACKGROUND: The presence of type 2 diabetes mellitus increases the risk of developing the colon cancer. The main objective of this study was to determine the role of sodium orthovanadate (SOV) in colon cancer associated with diabetes mellitus by targeting the competitive inhibition of PTP1B. METHODS: For in vivo study, high fat diet with low dose streptozotocin model was used for inducing the diabetes mellitus. Colon cancer was induced by injecting 1,2-dimethylhydrazine (25 mg/kg, sc) twice a week. TNM staging and immunohistochemistry (IHC) was carried out for colon cancer tissues. In vitro studies like MTT assay, clonogenic assay, rhodamine-123 dye assay and annexin V-FITC assay using flow cytometry were performed on HCT-116 cell line. CAM assay was performed to examine the anti-angiogenic effect of the drug. RESULTS: Sodium orthovanadate reduces the blood glucose level and tumor parameters in the animals. In vitro studies revealed that SOV decreased cell proliferation dose dependently. In addition, SOV induced apoptosis as depicted from rhodamine-123 dye assay and annexin V-FITC assay using flow cytometry as well as p53 IHC staining. SOV showed reduced angiogenesis effect on eggs which was depicted from CAM assay and also from CD34 and E-cadherin IHC staining. CONCLUSIONS: Our data suggest that SOV exhibits protective role in colon cancer associated with diabetes mellitus. SOV exhibits anti-proliferative, anti-angiogenic and apoptotic inducing effects hence can be considered for therapeutic switching in diabetic colon cancer.


Assuntos
Neoplasias do Colo , Diabetes Mellitus Tipo 2 , Animais , Glicemia , Vanadatos/farmacologia , Vanadatos/uso terapêutico , Neoplasias do Colo/patologia , Apoptose , Rodaminas/farmacologia , Rodaminas/uso terapêutico
8.
J Control Release ; 365: 1074-1088, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38101752

RESUMO

Oxidative stress is a major obstacle for neurological functional recovery after hypoxia-ischemia (HI) brain damage. Nanozymes with robust anti-oxidative stress properties offer a therapeutic option for HI injury. However, insufficiency of nanozyme accumulation in the HI brain by noninvasive administration hinders their application. Herein, we reported a cerium vanadate (CeVO4) nanozyme to realize a noninvasive therapy for HI brain in neonatal mice by targeting brain neuron mitochondria. CeVO4 nanozyme with superoxide dismutase activity mainly co-located with neuronal mitochondria 1 h after administration. Pre- and post-HI administrations of CeVO4 nanozyme were able to attenuate acute brain injury, by inhibiting caspase-3 activation, microglia activation, and proinflammation cytokine production in the lesioned cortex 2 d after HI injury. Moreover, CeVO4 nanozyme administration led to short- and long-term functional recovery following HI insult without any potential toxicities in peripheral organs of mice even after prolonged delivery for 4 weeks. These beneficial effects of CeVO4 nanozyme were associated with suppressed oxidative stress and up-regulated nuclear factor erythroid-2-related factor 2 (Nrf2) expression. Finally, we found that Nrf2 inhibition with ML385 abolished the protective effects of CeVO4 nanozyme on HI injury. Collectively, this strategy may provide an applicative perspective for CeVO4 nanozyme therapy in HI brain damage via noninvasive delivery.


Assuntos
Hipóxia-Isquemia Encefálica , Vanadatos , Animais , Camundongos , Animais Recém-Nascidos , Vanadatos/uso terapêutico , Vanadatos/metabolismo , Vanadatos/farmacologia , Administração Intranasal , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Fator 2 Relacionado a NF-E2/uso terapêutico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Isquemia/tratamento farmacológico , Mitocôndrias
9.
Nanotechnology ; 24(6): 065101, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23324398

RESUMO

Polyol mediated synthesized luminescent YVO(4):Eu(3+) nanoparticles (NPs) have been encapsulated in mesoporous silica nanoparticles (MSNs) using the sol-gel process. X-ray diffraction and Fourier transform infrared spectroscopy along with transmission electron microscopy confirm the encapsulation of the YVO(4):Eu(3+) NPs in the SiO(2) matrix. N(2) adsorption/desorption analysis confirms the mesoporous nature of the MSNs and YVO(4):Eu(3+)-MSNs. No significant quenching of the YVO(4):Eu(3+) luminescence is observed for YVO(4):Eu(3+)-MSNs. This nanocomposite has been tested as a potential drug carrier. Efficient loading of doxorubicin hydrochloride (DOX), a typical anticancer drug, is observed which reaches up to 93% in 8 mg ml(-1) of YVO(4):Eu(3+)-MSNs. pH sensitive release of DOX is observed, with 54% release for pH 4.3 and 31% in a physiological environment (pH 7.4). Both MSNs and YVO(4):Eu(3+)-MSNs nanocomposites do not show accountable toxicity to two cell lines, i.e. HeLa and MCF-7. However, as desired, toxicity is observed when cells are incubated with DOX loaded YVO(4):Eu(3+)-MSNs. Laser scanning confocal microscopy images confirm the uptake of the nanocomposite in both cell lines. The morphology of the cells (MCF-7) changes after incubation with DOX loaded YVO(4):Eu(3+)-MSNs, indicating an interaction of DOX with the cells. More cytotoxicity to both cell lines with ∼90% killing is observed due to the synergistic effect of magnetic fluid hyperthermia and chemotherapy using a biphasic suspension of superparamagnetic iron oxide magnetic nanoparticles and DOX loaded YVO(4):Eu(3+)-MSNs. In addition, an AC magnetic field triggers an enhanced drug release.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Nanopartículas/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/terapia , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Portadores de Fármacos/toxicidade , Európio/química , Európio/uso terapêutico , Európio/toxicidade , Óxido Ferroso-Férrico/química , Óxido Ferroso-Férrico/uso terapêutico , Óxido Ferroso-Férrico/toxicidade , Humanos , Hipertermia Induzida/métodos , Magnetismo/métodos , Nanopartículas/química , Nanopartículas/toxicidade , Dióxido de Silício/química , Dióxido de Silício/uso terapêutico , Dióxido de Silício/toxicidade , Vanadatos/química , Vanadatos/uso terapêutico , Vanadatos/toxicidade , Ítrio/química , Ítrio/uso terapêutico , Ítrio/toxicidade
10.
J Mater Sci Mater Med ; 24(4): 911-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23397279

RESUMO

The scope of this work was to synthesize a novel bifunctionalized vanadyl(VO)(2+)/vitamin B 6 complex. The diabetic therapeutic efficacy of the new complex was investigated in alloxan-induced diabetic mice. The results suggested that vanadyl(VO)(2+)/vit B6 complex has an anti-diabetic potency, improved the lipid profile and liver and kidney functions. The new complex possesses an antioxidant activity. The current results support the therapeutic potentiality of vanadyl(VO)(2+)/vitamin B 6 complex for the management of diabetes.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Vanadatos/uso terapêutico , Vitamina B 6/uso terapêutico , Aloxano , Animais , Creatinina/sangue , Teste de Tolerância a Glucose , Glucosefosfato Desidrogenase/metabolismo , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Ácido Úrico/sangue
11.
ACS Nano ; 17(17): 17105-17121, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37603593

RESUMO

Bioactive inorganic nanomaterials and the biological effects of metal ions have attracted extensive attention in tumor therapy in recent years. Vanadium (V), as a typical bioactive metal element, regulates a variety of biological functions. However, its role in antitumor therapy remains to be revealed. Herein, biodegradable vanadium disulfide (VS2) nanosheets (NSs) were prepared as a responsive gas donor and bioactive V source for activating cancer immunotherapy in combination with immune-checkpoint blockade therapy. After PEGylation, VS2-PEG exhibited efficient glutathione (GSH) depletion and GSH-activated hydrogen sulfide (H2S) release. Exogenous H2S caused lysosome escape and reduced adenosine triphosphate (ATP) synthesis in tumor cells by interfering with the mitochondrial membrane potential and inducing acidosis. In addition, VS2-PEG degraded into high-valent vanadate, leading to Na+/K+ ATPase inhibition, potassium efflux, and interleukin (IL)-1ß production. Together with further induction of ferroptosis and immunogenic cell death, a strong antitumor immune response was stimulated by reversing the immunosuppressive tumor microenvironment. Moreover, the combined treatment of VS2-PEG and α-PD-1 amplified antitumor therapy, significantly suppressed tumor growth, and further elicited robust immunity to effectively defeat tumors. This work highlights the biological effects of vanadium for application in cancer treatment.


Assuntos
Neoplasias , Vanadatos , Vanadatos/farmacologia , Vanadatos/uso terapêutico , Inibidores de Checkpoint Imunológico , Vanádio , Imunoterapia , Glutationa , Neoplasias/tratamento farmacológico
12.
J Neurosci Res ; 90(3): 691-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22183833

RESUMO

Attenuation of blood-brain barrier (BBB) disruption is one of the therapeutic candidates for treatment of subarachnoid hemorrhage (SAH). In this study, the protective effect of sodium orthovanadate (SOV) on BBB disruption was investigated in SAH using the endovascular perforation model. Fifty-five rats were randomly assigned to sham-operated, SAH treated with saline (as a vehicle), or 10 mg/kg SOV groups and were evaluated for neurofunction and Evans blue dye extravasation. The phosphorylation of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and mitogen-activated protein kinase (MAPK) and the expression of matrix metalloproteinase-9 (MMP-9), occludin, and collagen-IV were examined by Western blot analyses. Cell death among endothelial cells was revealed by immunofluorescence and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. SOV significantly improved neurofunction and reduced Evans blue dye extravasation in brains after SAH. SOV phosphorylated PTEN, decreased phospho-JNK and MMP-9, and preserved occludin expression. SOV also attenuated SAH-induced capillary endothelial cell death. The current study showed that SOV was protective against BBB disruption after SAH, possibly via PTEN phosphorylation.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , PTEN Fosfo-Hidrolase/metabolismo , Hemorragia Subaracnóidea/tratamento farmacológico , Vanadatos/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Morte Celular/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ocludina , Permeabilidade/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia , Vanadatos/uso terapêutico
13.
Invest New Drugs ; 30(4): 1302-10, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21553345

RESUMO

Gliomas are the most common primary brain tumor, and their treatment is still a challenge. Here, we evaluated the antiproliferative effect of a novel combination of two potent oxidative stress enhancers: menadione (M) and sodium orthovanadate (SO). We observed both short-term and prolonged growth inhibitory effects of M or SO alone as well as in combination (M:SO) on DBTRG.05MG human glioma cells. A stronger antiproliferative effect was observed in the short-term proliferation assay with the M:SO combination compared to either investigated agent alone. In the long-term proliferation assay, a 10-day exposure to M:SO at concentrations of 10 µM:17.5 µM or 17.5 µM:10 µM was enough to kill 100% of the cells; no cell regrowth was observed after re-incubation in drug-free media. When used in combination, the single concentration of M and SO could be decreased by 2.5- to 5-fold of those used for each experimental drug alone and still obtain a similar antiproliferative effect. The underlying molecular mechanism was investigated by co-incubating M:SO with dithiothreitol (DTT) and genistein. Both substances partially neutralized the effects of the M:SO combination, showing additive effects. This observation suggests a role of oxidative stress and tyrosine kinase stimulation in the M:SO cytotoxic effect. Our results indicate that M:SO combination is an attractive alternative for glioma treatment that encourages further study. The neutralizing effects of genistein and DTT reveal a possibility for their use in the minimization of potential M:SO systemic toxicity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Vanadatos/uso terapêutico , Vitamina K 3/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ditiotreitol/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Genisteína/farmacologia , Humanos , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Fatores de Tempo , Vanadatos/farmacologia , Vitamina K 3/farmacologia
14.
Eur J Pharmacol ; 919: 174798, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35123977

RESUMO

Depression is a psychiatric disorder characterized by low-esteem, anhedonia, social deficit, and lack of interest. Decreased brain-derived neurotrophic factor (BDNF) and impaired tropomyosin kinase B receptor (TrkB receptor) signaling are associated with depression. In our study, depressive-like behavior was induced in mice by chronic unpredictable mild stress (CUMS) model. Various behavioral tests like tail suspension test (TST), open field test (OFT), sucrose preference test (SPT); biochemical analyses for corticosterone, reduced glutathione (GSH), lipid peroxidation (LPO), superoxide dismutase (SOD), nitric oxide (NO) and enzyme-linked immunosorbent assay (ELISA) for BDNF were performed. Body weight was measured every week. CUMS induced depressive-like behavior was found to be associated with increased oxidative stress in the brain and serum corticisterone with subsequent reduction of BDNF. Sodium orthovanadate (SOV), a protein tyrosine phosphatase inhibitor already reported to elevate BDNF levels, was used as the test drug. Sodium orthovanadate (5 mg/kg, 10 mg/kg) and fluoxetine (FLX-10 mg/kg) was given to mice orally for 21days before 30 min of stress induction. The behavioral tests reflected depressive-like behavior in CUMS, which was attenuated by both SOV and fluoxetine. SOV at 10 mg/kg demonstrated significant results in the present study characterized by decreased malondialdehyde levels (MDA/LPO), NO levels, and increased GSH level and SOD activity in both the cortex and hippocampus. Besides, ELISA has revealed the significant elevation of BDNF levels in the treatment groups (SOV-5 mg/kg, 10 mg/kg and FLX-10 mg/kg) as compared to the disease group (CUMS). Therefore, the treatment with SOV appeared to reverse both oxidative and nitrosative stress. Decreased serum corticosterone levels observed with SOV (5 & 10 mg/kg), FLX-10 mg/kg, FLX (10 mg/kg) + SOV (5 mg/kg); and SOV-10 mg/kg per-se treatment and elevated BDNF level with SOV (5 & 10 mg/kg), FLX-10 mg/kg were associated with attenuation of depressive-like behavior. The findings of this preliminary study indicate that SOV has the potential to restore antidepressant-like effects or prevent stress-induced anhedonia and so further molecular mechanisms are warranted for clinical translation.


Assuntos
Antidepressivos/farmacologia , Estresse Psicológico , Vanadatos/farmacologia , Administração Oral , Animais , Antidepressivos/química , Antidepressivos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Vanadatos/química , Vanadatos/uso terapêutico
15.
Stroke ; 42(2): 477-83, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21193742

RESUMO

BACKGROUND AND PURPOSE: Recent studies reported that apoptosis was involved in the pathogenesis of early brain injury after subarachnoid hemorrhage (SAH). The aim of this study was to examine whether sodium orthovanadate (SOV) prevents post-SAH apoptosis by modulating growth factors and its downstream receptor tyrosine kinases. Method- Rats were operated on with the endovascular perforation model. SAH animals were treated with vehicle, 3 mg/kg and 10 mg/kg SOV, and evaluated regarding neurofunction and brain edema. The expression of growth factors such as mature brain-derived neurotrophic factor, insulin-like growth factor-1, and vascular endothelial growth factor and phosphorylation of tropomyosin-related kinase B, which is a receptor tyrosine kinase for brain-derived neurotrophic factor and the downstream pathway in antiapoptosis, was examined by Western blot analysis. Neuronal cell death was measured with terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling staining. We also administered K252a, a tropomyosin-related kinase B antagonist, to examine the mechanisms for neuroprotective effects by SOV. RESULTS: SOV significantly improved neurofunction and reduced brain edema after SAH. SOV increased mature brain-derived neurotrophic factor and prevented post-SAH tropomyosin-related kinase B inactivation and caspase-3 activation, resulting in attenuation of neuronal cell death in the cortex and hippocampal CA1 region. Preinjection of K252a abolished the beneficial effects of SOV. CONCLUSIONS: The current study showed that brain-derived neurotrophic factor-induced tropomyosin-related kinase B activation by SOV was necessary for protection against early brain injury after SAH.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor trkB/fisiologia , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/enzimologia , Tropomiosina/fisiologia , Vanadatos/uso terapêutico , Animais , Lesões Encefálicas/enzimologia , Lesões Encefálicas/etiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Hemorragia Subaracnóidea/complicações , Fatores de Tempo , Vanadatos/farmacologia
16.
Biometals ; 24(6): 1153-68, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21755303

RESUMO

The complex of vanadyl(IV) cation with oxodiacetate, VO(oda) caused an inhibitory effect on the proliferation of the human colon adenocarcinoma cell line Caco-2 in the range of 25-100 µM (P < 0.001). This inhibition was partially reversed by scavengers of free radicals. The difference in cell proliferation in the presence and the absence of scavengers was statistically significant in the range of 50-100 µM (P < 0.05). VO(oda) altered lysosomal and mitochondria metabolisms (neutral red and MTT bioassays) in a dose-response manner from 10 µM (P < 0.001). Morphological studies showed important transformations that correlated with the disassembly of actin filaments and a decrease in the number of cells in a dose response manner. Moreover, VO(oda) caused statistically significant genotoxic effects on Caco-2 cells in the low range of concentration (5-25 µM) (Comet assay). Increment in the oxidative stress and a decrease in the GSH level are the main cytotoxic mechanisms of VO(oda). These effects were partially reversed by scavengers of free radicals in the range of 50-100 µM (P < 0.05). Besides, VO(oda) interacted with plasmidic DNA causing single and double strand cleavage, probably through the action of free radical species. Altogether, these results suggest that VO(oda) is a good candidate to be evaluated for alternative therapeutics in cancer treatment.


Assuntos
Acetatos/toxicidade , Acetatos/uso terapêutico , Células CACO-2/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Vanadatos/toxicidade , Vanadatos/uso terapêutico , Acetatos/química , Actinas/metabolismo , Animais , Células CACO-2/citologia , Proliferação de Células/efeitos dos fármacos , Ensaio Cometa , Citoesqueleto/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Vanadatos/química
17.
Acta Neurochir Suppl ; 110(Pt 1): 67-70, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116917

RESUMO

PURPOSE: Sodium orthovanadate (SOV) is a representative tyrosine phosphatase inhibitor and has been shown to ameliorate neuronal injury in cerebral ischemia. We hypothesized that tyrosine phosphatase inhibition by SOV might attenuate early brain injury after subarachnoid hemorrhage (SAH) in this study. METHODS: The endovascular perforation model of SAH was produced and animals were randomly assigned to sham-operated rats, saline-treated (vehicle), and 10 mg/kg of SOV-treated SAH rats. Drugs were injected intraperitoneally immediately after SAH induction. Neurological score and brain water content (BWC) were assessed at 24 h after SAH. Cell injury was studied by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL) at 24 h after SAH. RESULTS: Severity of SAH and mortality in SOV-treated rats was similar to that of the saline group. SOV significantly decreased BWC and improved neurological score at 24 h after SAH compared with the saline group. SOV decreased TUNEL-positive cells at 24 h after SAH compared with the saline group. CONCLUSIONS: These data suggest that tyrosine phosphatase inhibition by SOV ameliorates early brain injury after SAH.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/enzimologia , Proteínas Tirosina Fosfatases/metabolismo , Vanadatos/uso terapêutico , Animais , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/mortalidade , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Exame Neurológico , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Hemorragia Subaracnóidea/complicações , Fatores de Tempo
18.
Biosci Rep ; 40(9)2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-32940655

RESUMO

Vanadium is an ultratrace element. The transition metal vanadium, widely exists in the environment and exhibits various biological and physiological effects in the human body, yet with no presently known specific physiological function in mammals. Sodium orthovanadate (SOV) is a kind of vanadium compound. SOV has shown promising antineoplastic activity in several human cancers. But the effects of SOV on acute promyelocytic leukemia (APL) are still unknown. In the present study, for the first time, we found that SOV could inhibit proliferation, induce G2/M cell cycle arrest and apoptosis, and could inhibit autophagy of acute leukemia cell lines in vitro. Thus, our findings suggest that SOV could effectively suppress the growth of acute leukemia HL60 cells and HL60/A cells through the regulations of proliferation, cell cycle, apoptosis and autophagy, and thus may act as a potential therapeutic agent in APL treatment.


Assuntos
Antineoplásicos/farmacologia , Leucemia Promielocítica Aguda/tratamento farmacológico , Vanadatos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/patologia , Vanadatos/uso terapêutico
19.
Biometals ; 22(6): 895-905, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19404749

RESUMO

Vanadium(III, IV, V)-chlorodipicolinate (dipic-Cl) complexes, including H[VIII(dipic-Cl)2] · 5H2O (V3dipic-Cl), VIVO(dipic-Cl)(H2O)2 (V4dipic-Cl) and K[VVO2(dipic-Cl)] (V5dipic-Cl), were prepared with the indicated oxidation states. Our aim was to evaluate the anti-diabetic effects of V3dipic-Cl, V4dipic-Cl and V5dipic-Cl in streptozotocin-induced diabetic rats. Vanadium complexes were orally administered to diabetic rats at concentrations of 0.1-0.3 mg/ml in the drinking water. We found that vanadium-chlorodipicolinate (V-dipic-Cl) complexes at the concentration of 0.1 mg/ml did not exhibit blood glucose-lowering effects when administered to diabetic rats for 20 days. However, the levels of fasting blood glucose in diabetic rats were decreased after treatment with 0.3 mg/ml of V4dipic-Cl and V5dipic-Cl complexes for the following 20 days. Although administration of both V4dipic-Cl and V5dipic-Cl significantly lowered diabetic hyperglycemia, the vanadium intake from administration of V4dipic-Cl is nearly 1.5-fold greater compared to that of V5dipic-Cl. Treatment with the H2dipic-Cl ligand and all three V-dipic-Cl complexes significantly lowered serum cholesterol, while administration of the V5dipic-Cl complex lowered serum cholesterol significantly more than administration of the ligand alone. Treatment with ligand alone did not have an effect on serum triglyceride, while administration of the V4dipic-Cl and V5dipic-Cl significantly lowered the elevated serum triglyceride associated with diabetes. Oral administration of the ligand and all V-dipic-Cl complexes did significantly lower diabetes elevated serum alkaline phosphatase. Treatment with H2dipic-Cl ligand and V4dipic-Cl and V5dipicCl significantly lowered diabetes elevated aspartate amino transferase. These results indicate that the health of the treated animals did not seem to be further compromised compared to that of diabetic animals. In addition, oral administration of H2dipic-Cl, V3dipic-Cl, V4dipic-Cl and V5dipic-Cl did not alter diabetic serum creatinine and blood urea nitrogen levels, suggesting no significant side effects of vanadium treatment on renal functions at the dose of 0.3 mg/ml in diabetic rats. The results presented here suggest that the anti-diabetic effects of treatment with V-dipic-Cl complexes were likely associated in part with the oxidation state of vanadium.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Vanadatos/uso terapêutico , Administração Oral , Fosfatase Alcalina/sangue , Animais , Aspartato Aminotransferases/sangue , Glicemia/análise , Nitrogênio da Ureia Sanguínea , Cloro/química , Colesterol/sangue , Creatinina/sangue , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/fisiopatologia , Modelos Animais de Doenças , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/sangue , Hipoglicemiantes/síntese química , Insulina/sangue , Testes de Função Renal , Oxirredução , Ácidos Picolínicos/química , Ratos , Ratos Wistar , Estreptozocina/toxicidade , Triglicerídeos/sangue , Vanadatos/administração & dosagem , Vanadatos/sangue , Vanadatos/síntese química , Vanádio/química
20.
Drug Des Devel Ther ; 13: 1409-1420, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118576

RESUMO

BACKGROUND: This study aimed to design a compound with folic acid (FAH2) and vanadyl (IV) for use in the treatment of diabetes. MATERIALS AND METHODS: A novel vanadyl (IV) FAH2 complex was synthesized and characterized [(FA2-)(VO2+)]⋅3H2O. The speculated structure of this folate complex was determined using physicochemical techniques including microanalytical analysis, conductivity studies, spectroscopic examination, magnetic measurements, thermogravimetric analyses, and morphological X-ray powder diffraction, and scanning and transmission electron microscopies. The anti-diabetic therapeutic potential of the complexes was tested in a 30-day streptozotocin-induced diabetes rat model. RESULTS: The conductivity test of the complex implied electrolyte behavior. The spectroscopic assessments of the isolated dark yellow solid complex revealed that FAH2 acts as a bidentate ligand. The coordination process with two vanadyl (IV) ions occurred through the deprotonation of both carboxyl groups of FAH2 in a regular square pyramid arrangement at a 2(FA)2-: 2(VO)2+ molar ratio. XRD, SEM, and TEM analyses revealed the complex crystalline nature of the complex. Treating diabetic rats with vanadyl (IV) FAH2 complex significantly improved many biological parameters relevant to diabetes pathology with minimal toxicity. CONCLUSION: The data generated in this study indicate that the synthesized vanadyl (IV) folate complex acts as a model of anti-diabetic agent.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Ácido Fólico/química , Hipoglicemiantes/química , Hipoglicemiantes/síntese química , Compostos Organometálicos/síntese química , Vanadatos/química , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Espectroscopia de Ressonância de Spin Eletrônica , Ácido Fólico/uso terapêutico , Hipoglicemiantes/uso terapêutico , Masculino , Estrutura Molecular , Compostos Organometálicos/química , Compostos Organometálicos/uso terapêutico , Tamanho da Partícula , Ratos , Espectrofotometria Infravermelho , Estreptozocina , Propriedades de Superfície , Termodinâmica , Vanadatos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA