Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Noppawan Phumala Morales is active.

Publication


Featured researches published by Noppawan Phumala Morales.


Journal of Ethnopharmacology | 2008

The in vitro and ex vivo antioxidant properties, hypolipidaemic and antiatherosclerotic activities of water extract of Moringa oleifera Lam. leaves

Pilaipark Chumark; Panya Khunawat; Yupin Sanvarinda; Srichan Phornchirasilp; Noppawan Phumala Morales; Laddawal Phivthong-ngam; Piyanee Ratanachamnong; Supath Srisawat; Klai-upsorn S. Pongrapeeporn

UNLABELLED Moringa oleifera is used in Thai traditional medicine as cardiotonic. Recent studies demonstrated its hypocholesterolaemic effect. However, to be clinically useful, more scientific data are needed. AIM OF THE STUDY We investigated the antioxidant, hypolipidaemic and antiatherosclerotic activities of Moringa oleifera leaf extract. MATERIALS AND METHODS Scavenging activity of the extract on 1,1-diphenyl-2-picrylhydrazyl radicals (DPPH), and the inhibitory effect on Cu(2+)-induced low-density lipoprotein (LDL) oxidation were determined in in vitro experiment. The effects of the extract on cholesterol levels, conjugated diene (CD) and thiobarbituric acid reactive substances (TBARS) and plaque formations in cholesterol-fed rabbits were investigated. RESULTS We found that in scavenging DPPH radicals the extract and Trolox had IC(50) of 78.15+/-0.92 and 2.14+/-0.12microg/ml, respectively. The extract significantly (P<0.05) prolonged the lag-time of CD formation and inhibited TBARS formation in both in vitro and ex vivo experiments in a dose-dependent manner. In hypercholesterol-fed rabbits, at 12 weeks of treatment, it significantly (P<0.05) lowered the cholesterol levels and reduced the atherosclerotic plaque formation to about 50 and 86%, respectively. These effects were at degrees comparable to those of simvastatin. CONCLUSIONS The results indicate that this plant possesses antioxidant, hypolipidaemic and antiatherosclerotic activities and has therapeutic potential for the prevention of cardiovascular diseases.


International Journal of Rheumatic Diseases | 2009

Lipid peroxidation, glutathione, vitamin E, and antioxidant enzymes in synovial fluid from patients with osteoarthritis.

Noppawan Phumala Morales; Keerati Charoencholvanich; Thossart Harnroongroj

Aim:  To compare levels of lipid peroxidation and antioxidants in synovial fluid from primary knee osteoarthritis (OA) patients with severe cartilage damage undergoing total knee replacement with those in the synovial fluid from injured knee joint patients with intact cartilage undergoing knee arthroscopy.


British Journal of Clinical Pharmacology | 2008

UGT1A6 genotype-related pharmacokinetics of deferiprone (L1) in healthy volunteers.

Lie Michael George Limenta; Totsapol Jirasomprasert; Jeeranut Tankanitlert; Saovaros Svasti; Prapin Wilairat; Udom Chantharaksri; Suthat Fucharoen; Noppawan Phumala Morales

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT UGT1A6 has been proposed as the predominant isoform responsible for the glucuronidation of deferiprone. UGT1A6*2 allele has been associated with the altered enzyme activity. WHAT THIS STUDY ADDS There is no statistically significant effect of UGT1A6 genotype on the single-dose pharmacokinetics of deferiprone in healthy volunteers. Gender influences serum pharmacokinetics of deferiprone. Body iron stores reflected by serum ferritin levels may have an influence on the extent of extravascular deferiprone distribution. AIMS To examine the effects of UGT1A6 polymorphisms on the pharmacokinetics of deferiprone in healthy volunteers. METHODS Twenty-two healthy volunteers were enrolled and grouped according to UGT1A6 genotype. After an overnight fast, the subjects received a single oral dose of 25 mg kg(-1) deferiprone. Blood samples were collected at 0, 15, 30, 45, 60, 90, 120, 180, 240, 300 and 360 min after dosing. Urine output was collected at 0, 0-2, 2-4, 4-8, 8-12 and 12-24 h. Deferiprone (L1) and deferiprone-glucuronide (L1G) concentrations in serum and urine were determined using a validated high-performance liquid chromatography method. UGT1A6 genotypes were determined by polymerase chain reaction-restriction fragment length polymorphism analysis. RESULTS No statistically significant differences in any pharmacokinetic parameters of either deferiprone or deferiprone-glucuronide among the genotype groups were noted. Likewise, there were no statistically significant differences in 24-h urinary deferiprone and deferiprone-glucuronide excretion among the genotype groups. Significant differences between men and women were found in AUC(0-infinity), V(d)/F, and CL/F of deferiprone. Gender differences in 24-h urinary deferiprone and its metabolite excretion, however, failed to reach statistical significance. The V(d)/F of deferiprone was found to correlate significantly with serum ferritin (r(s) = 0.665; P = 0.001). CONCLUSION The studied single nucleotide polymorphisms in UGT1A6 do not appear to exert statistically significant effects on the single-dose pharmacokinetics of deferiprone. Gender appears to influence the serum pharmacokinetics of deferiprone, but not urinary excretion of deferiprone and its metabolite. Body iron stores may have an influence on the extent of extravascular deferiprone distribution.


Neuroscience Letters | 2011

Caffeine potentiates methamphetamine-induced toxicity both in vitro and in vivo.

Theerin Sinchai; Surin Plasen; Yupin Sanvarinda; Yamaratee Jaisin; Piyarat Govitrapong; Noppawan Phumala Morales; Piyanee Ratanachamnong; Duangporn Plasen

Ya-Ba, a combination of the two potent psychostimulants methamphetamine (METH) and caffeine (CAF), is commonly used by drug abusers in Thailand and neighboring countries. While the neurotoxic effects of METH are well documented, the toxicity of this combination is mostly unknown. This study aimed to elucidate the effects of this particular drug combination using both in vitro and in vivo models. We found that combined treatment of METH and CAF at individually non-toxic concentrations significantly decreased viability of human neuroblastoma SK-N-SH cells. The reduction in cell survival was accompanied by an increase in reactive oxygen species (ROS) formation and the Bax/Bcl-2 ratio. In vivo data showed that combined administration of METH and CAF increased the mortality rate of rats, with an increase in the level of thiobarbituric acid reactive substances (TBARS), the indicator of oxidative stress, in striatal tissues. The results indicate that caffeine potentiates the toxic effects of methamphetamine, possibly via a mechanism involving an increase in dopamine release and excess ROS generation.


Pharmacology | 2007

Effects of Combined UDP-Glucuronosyltransferase (UGT) 1A1*28 and 1A6*2 on Paracetamol Pharmacokinetics in β-Thalassemia/HbE

Jeeranut Tankanitlert; Noppawan Phumala Morales; Thad A. Howard; Pranee Fucharoen; Russell E. Ware; Suthat Fucharoen; Udom Chantharaksri

In addition to pathophysiological changes, genetic variations can alter drug pharmacokinetics in patients with thalassemia. Numerous drugs are metabolized by UDP-glucuronosyltransferases (UGT) including paracetamol (PCM), a widely used analgesic. Co-occurrence of the UGT1A1 polymorphism (UGT1A1*28) and the UGT1A6 polymorphism (UGT1A6*2) may affect PCM glucuronidation. To elucidate the effect of these combined polymorphisms on the PCM metabolism in thalassemic patients, 15 β-thalassemia/hemoglobin E subjects with three different UGT1A genotypes received a single oral dose of 1,000 mg PCM. Drug disposition was determined by HPLC. Patients who have UGT1A6*2 without UGT1A1*28 showed a significant, lower area under concentration-time curve (AUC₀→∞) of PCM, PCM-glucuronide and PCM-sulfate than those of the patients with wild-type UGT1A1 and UGT1A6 (p < 0.05). In addition, a high elimination rate constant and clearance of PCM and its metabolites were also found in these patients (p < 0.05). Ourstudy suggests that a subtherapeutic level of PCM may occur in patients who have UGT1A6*2 without UGT1A1*28.


Free Radical Biology and Medicine | 2012

Whole-body kinetic image of a redox probe in mice using Overhauser-enhanced MRI.

Nuttavut Kosem; Tatsuya Naganuma; Kazuhiro Ichikawa; Noppawan Phumala Morales; Keiji Yasukawa; Fuminori Hyodo; Ken-ichi Yamada; Hideo Utsumi

Overhauser-enhanced MRI (OMRI) enables visualization of free radicals in animals based on dynamic nuclear polarization. Real-time data of tissue redox status gathered from kinetic images of redox-sensitive nitroxyl radical probes using OMRI provided both anatomic and physiological information. Phantom experiments demonstrated the linear correlation between the enhancement factor and the concentration of a membrane-impermeable probe, carboxy-PROXYL (3-carboxy-2,2,5,5-tetramethyl- pyrrolidine-1-oxyl). Whole-body OMRI images illustrated the in vivo kinetics of carboxy-PROXYL for 25 min. Initial distribution was observed in lung, heart, liver, and kidney, but not brain, corresponding to its minimal lipophilicity. Based on these images (pixel size, 1.33 × 1.33 mm; slice thickness, 50mm), a time-concentration curve with low coefficient of variance (<0.21) was created to assess pharmacokinetic behaviors. A biexponential curve showed a distribution phase from 1 to 10 min and an elimination phase from 15 to 25 min. The α rate constant was greater than the β rate constant in ROIs, confirming that its pharmacokinetics obeyed a two-compartment model. As a noninvasive technique, combining OMRI imaging with redox probes to monitor tissue redox status may be useful in acquiring valuable information regarding organ function for preclinical and clinical studies of oxidative diseases.


Phytotherapy Research | 2014

Curcumin I Mediates Neuroprotective Effect Through Attenuation of Quinoprotein Formation, p‐p38 MAPK Expression, and Caspase‐3 Activation in 6‐Hydroxydopamine Treated SH‐SY5Y Cells

Benjawan Meesarapee; Anusorn Thampithak; Yamaratee Jaisin; Pimtip Sanvarinda; Apichart Suksamrarn; Patoomratana Tuchinda; Noppawan Phumala Morales; Yupin Sanvarinda

6‐Hydroxydopamine (6‐OHDA) selectively enters dopaminergic neurons and undergoes auto‐oxidation resulting in the generation of reactive oxygen species and dopamine quinones, subsequently leading to apoptosis. This mechanism mimics the pathogenesis of Parkinsons disease and has been used to induce experimental Parkinsonism in both in vitro and in vivo systems. In this study, we investigated the effects of curcumin I (diferuloylmethane) purified from Curcuma longa on quinoprotein production, phosphorylation of p38 MAPK (p‐p38), and caspase‐3 activation in 6‐OHDA‐treated SH‐SY5Y dopaminergic cells. Pretreatment of SH‐SY5Y with curcumin I at concentrations of 1, 5, 10, and 20 μM, significantly decreased the formation of quinoprotein and reduced the levels of p‐p38 and cleaved caspase‐3 in a dose‐dependent manner. Moreover, the levels of the dopaminergic neuron marker, phospho‐tyrosine hydroxylase (p‐TH), were also dose‐dependently increased upon treatment with curcumin I. Our results clearly demonstrated that curcumin I protects neurons against oxidative damage, as shown by attenuation of p‐p38 expression, caspase‐3‐activation, and toxic quinoprotein formation, together with the restoration of p‐TH levels. This study provides evidence for the therapeutic potential of curcumin I in the chemoprevention of oxidative stress‐related neurodegeneration. Copyright


Clinical Pharmacokinectics | 2011

Pharmacokinetics of deferiprone in patients with β-thalassaemia: impact of splenectomy and iron status.

Lie Michael George Limenta; Totsapol Jirasomprasert; Piyada Jittangprasert; Prapin Wilairat; Praveena Yamanont; Udom Chantharaksri; Suthat Fucharoen; Noppawan Phumala Morales

Background and ObjectiveIron-rich transfusions and/or a compensatory increase in iron absorption ultimately result in iron loading in patients with β-thalassaemia. Hence, without iron chelation, iron accumulates relentlessly. Deferiprone has been shown to be capable of reducing the iron burden in patients with b-thalassaemia. However, there is wide interpatient variation in deferiprone-induced urinary iron excretion (UIE). We hypothesized that splenectomy and iron status might influence the pharmacokinetic profiles of deferiprone in patients with β-thalassaemia/haemoglobin E, and the present study was aimed at examining this hypothesis.Study Participants and MethodsThirty-one patients with β-thalassaemia/haemoglobin E (20 splenecto-mized and 11 non-splenectomized patients) were enrolled in the study. After an overnight fast, the subjects received a single oral dose of deferiprone 25 mg/kg of body weight. Blood samples were collected pre-dosing and at 15, 30, 45, 60, 90, 120, 180, 240, 300, 360 and 480 minutes after dosing. Urine output was pooled and collected at 0–2, 2–4, 4–8, 8–12 and 12–24 hour intervals. Serum and urine concentrations of deferiprone and its metabolite deferiprone glucuronide were determined using a validated high-performance liquid chromatography method. Serum deferiprone-chelated iron and UIE were determined using a validated colourimetric method.ResultsNo significant difference in the pharmacokinetic parameters of non-conjugated deferiprone was observed between splenectomized and non-splenectomized patients. However, the maximum serum concentration (Cmax) and the area under the serum concentration-time curve (AUC) from time zero to infinity (AUC∞) values of deferiprone glucuronide were significantly lower (both p < 0.05) in splenectomized patients (median 53.2µmol/L and 12 634 µmol · min/L, respectively) than in non-splenectomized patients (median 70.5 µmol/L and 20 601 mmol · min/L, respectively). The Cmax and the AUC from time zero to the time of the last measurable concentration (AUClast) values of serum deferiprone-chelated iron, as well as UIE, were significantly higher (p < 0.001) in splenectomized patients (median values 7.1 µmol/L, 1645 mmol · min/L and 77.1 mmol, respectively) than in non-splenectomized patients (median values 3.1 µmol/L, 545 mmol · min/L and 12.5 µmol, respectively). Urinary excretion of non-conjugated deferiprone and deferiprone glucuronide did not differ between the two groups. Further analyses using multiple linear regressions indicated that the iron profiles (non-transferrin-bound iron and ferritin) were significant predictors of the pharmacokinetic parameters of non-conjugated deferiprone, deferiprone-chelated iron and UIE. In addition, splenectomy status was identified as the strongest predictor of the AUClast of deferiprone-chelated iron and UIE.ConclusionBoth iron and splenectomy status have significant effects on the pharmacokinetics and iron chelation efficacy of deferiprone. A greater degree of iron overload in splenectomized patients results in alterations in pharmacokinetic parameters (the Cmax and AUC) of deferiprone glucuronide and deferiprone-chelated iron, as well as a significant increase in UIE.


Free Radical Research | 2009

Pharmaco/ferrokinetic-related pro-oxidant activity of deferiprone in β-thalassemia

Totsapol Jirasomprasert; Noppawan Phumala Morales; Lie Michael George Limenta; Paveena Yamanont; Prapin Wilairat; Suthat Fucharoen; Udom Chantharaksri

The potential of free radical formation in serum of β-thalassemia/Hb E patients receiving a single oral dose of 25 mg/kg body weight of deferiprone, a bidentate orally active iron chelator, was evaluated using EPR/spin trapping technique. In the presence of ascorbic acid and tert-butylhydroperoxide, EPR signals of ascorbyl radical (aH=0.18 mT) and DMPO-carbon centred adduct (aH=2.37 mT, aN=1.65 mT) were detected. Shortly after deferiprone administration, EPR signal intensities decreased concomitant with an increase in serum levels of deferiprone. Unfortunately, enhanced EPR signal intensities were observed at 300 min after dosing in patients with serum molar ratio of deferiprone to iron less than 3, suggesting the formation of incomplete iron-deferiprone complexes and consequently free radical formation. To avoid adverse effects of deferiprone, a dosage regimen should be designed according to iron status of the patients and aimed at maintaining an adequate ratio of serum chelator-to-iron concentration.


Clinical Chemistry and Laboratory Medicine | 2006

The reduction of cholesteryl linoleate in lipoproteins: an index of clinical severity in β-thalassemia/Hb E

Rataya Luechapudiporn; Noppawan Phumala Morales; Suthat Fucharoen; Udom Chantharaksri

Abstract Background: Oxidative modification of lipoproteins has been reported in β-thalassemia and has been suggested to relate to atherogenesis-risk. This study focused on the change in cholesteryl esters in plasma lipoproteins under oxidative stress resulting from iron overload in β-thalassemia/hemoglobin E (β-thal/Hb E) patients. Methods: Markers of oxidative damage and cholesteryl esters (CEs) were measured in plasma and lipo-proteins from 30 β-thal/Hb E patients and compared to those from 10 healthy volunteers. CEs in plasma, low-density lipoprotein (LDL) and high-density lipoprotein (HDL) were separated and identified using HPLC. Results: β-Thal/Hb E patients presented iron overload, a precipitous decrease in α-tocopherol and increased lipid peroxidation (thiobarbituric acid-reactive substances; TBARs) in both plasma and lipoproteins. Cholesteryl linoleate, the most abundant CE in lipoproteins, showed a reduction of 70% in LDL, while other CEs showed a lower reduction (50%). An inverse relationship between the cholesteryl linoleate/cholesteryl oleate ratio (CL/CO) and the degree of clinical severity suggested that the CL/CO ratio is an index of damaged lipoproteins and could be used as a pathologic marker of underlying iron overload. Good correlation of non-transferrin-bound iron (NTBI) and TBARs (r=0.8, p<0.01) in LDL strongly supported the contention that iron overload is responsible for initiating the lipid peroxidation in β-thal/Hb E. Conclusions: This study suggests that cholesteryl linoleate is the primary target of oxidative modification induced by NTBI in β-thal/Hb E patients and that reduction in cholesteryl linoleate in lipoproteins could be used as a severity index for β-thal/Hb E.

Collaboration


Dive into the Noppawan Phumala Morales's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hamady Dieng

Universiti Malaysia Sarawak

View shared research outputs
Top Co-Authors

Avatar

Fatimah Abang

Universiti Malaysia Sarawak

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge