Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Cureus ; 15(9): e44621, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37799254

ABSTRACT

Background Serum prostate-specific antigen (PSA) is a well-established marker that can be measured as an indicator for screening, diagnosing, and managing prostate cancer due to its advanced tissue specificity. Numerous studies have revealed that free PSA is the predominant molecular form of PSA in breast cancer cases. In contrast, total PSA is prevalent in benign breast tumor cases and healthy females. This case-control study aims to measure PSA levels among individuals with breast cancer in order to establish PSA as a prognostic biomarker. Methods The study involved 150 female subjects between the ages of 18 and 70 and was conducted between 2013 and 2014. The subjects were then categorized into three groups: those with malignant breast cancer, those with benign breast tumors, and the control group with no history of malignant or benign breast tumors. Participants were asked to complete a lifestyle questionnaire and interview using hospital medical records to establish past and pertinent patient medical history. These cases were acquired from the 7th of October Hospital's surgery department and Benghazi Central Hospital's oncology clinic in Libya. Sandwich-type ELISA's were used for PSA quantitation, while the Wilcoxon Rank-Sum test was used to identify statistically significant differences between total PSA and free PSA measurements within each patient group. Results This study did not reveal significant statistical differences in total PSA levels between breast cancer cases and control groups (p=0.200), or between breast cancer and fibroadenoma patients (p=0.472). However, there was a significant difference in F-PSA levels between breast cancer and fibroadenoma cases (p=0.0001). Neither total-PSA (p=0.200) nor F-PSA (p=0.262) levels showed significant differences between breast cancer cases and controls. This study paved the way for further investigations into PSA's role in breast cancer. Despite its limitations, it offers an opportunity to delve deeper into understanding PSA's potential role and use in breast cancer. Conclusion A comprehensive statistical analysis revealed a positive correlation between F-PSA levels and breast cancer diagnosis. The findings suggest that PSA may serve as a prognostic biomarker for breast cancer. This may contribute to improved customized treatment approaches, offering precise and accurate risk assessments, understanding breast cancer biology, and improving health outcomes for patients with breast cancer.

2.
Cureus ; 15(1): e33648, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36788860

ABSTRACT

Cardiovascular diseases are one of the leading causes of death in the United States; therefore, primary and secondary prevention are of the utmost importance. In this regard, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMG-CoA) reductase inhibitors, also known as statins, have been anointed as the primary treatment method for lowering cholesterol to prevent cardiovascular diseases. Statins decrease the low-density lipoprotein (LDL) cholesterol and triglycerides in the body, thus lowering the total body cholesterol levels. Despite the benefits associated with statins, it is essential to understand the adverse effects of these drugs. Myotoxicity and statin-associated muscle symptoms are the most common adverse effects of statins. The impairment of mitochondrial function is another adverse effect that can lead to hepatic dysfunction, neurocognitive effects, and potentially the new onset of diabetes. The exact pathophysiology of these side effects is still not fully understood. However, several mechanisms have been proposed, although there is significant overlap among the hypothetical propositions. Understanding the overall outcomes of each of these adverse effects can allow a healthcare practitioner to carefully map out whether statin administration should be used to prevent hypercholesterolemia in the body. The adverse effect of statins is dependent on both the dose and the type of statin used. Lipophilic statins tend to possess a more remarkable ability to infiltrate membranes; they have been hypothesized to cause statin-induced myopathies as well as neurocognitive effects by significantly crossing the blood-brain barrier. In summary, this review has focused on the mechanistic and clinical aspects of this statin class of medication. Proposed mechanisms for different adverse effects associated with statins remain a focus of this communication.

3.
Am J Pharm Educ ; 86(5): 8764, 2022 06.
Article in English | MEDLINE | ID: mdl-34507961

ABSTRACT

Objective To explore methods that pharmacy programs can use to redefine their work environment to reduce stress, improve well-being, and increase faculty productivity.Findings To demonstrate a culture of support, organizations should consider a five-fold approach to enhancing and maintaining faculty well-being, including optimizing faculty and staff support, establishing a faculty development and mentoring program, permitting flexibility in work schedules, improving productivity of meetings, and managing communication tools. Individuals can also take measures to improve their well-being, including controlling email, giving attention to faculty citizenship, implementing stress reduction and coping techniques, and maintaining boundaries between work and home.Summary This article discusses approaches that have been shown to reduce burnout and provides strategies organizations and individuals can implement to improve productivity and faculty well-being. While certain areas, such as faculty wellness and productivity, have been well-studied in the pharmacy and health professions literature, significant gaps were identified in other areas, including alternate work arrangements. In some cases, data from the business sector can be extrapolated to pharmacy education; however, inferences from effective corporate strategies may not be transferable to the culture and expectations of academia. While there is significant overlap between institutional and individual strategies, a culture of communication, collaboration, support, and citizenship is foundational. There is no single strategy that will work for everyone, and flexibility is important to develop an individualized approach.


Subject(s)
Burnout, Professional , Education, Pharmacy , Mentoring , Burnout, Professional/prevention & control , Faculty , Faculty, Pharmacy , Humans
4.
Molecules ; 25(6)2020 Mar 19.
Article in English | MEDLINE | ID: mdl-32204372

ABSTRACT

Curcumin exerts a wide range of beneficial physiological and pharmacological activities, including antioxidant, anti-amyloid, anti-inflammatory, anti-microbial, anti-neoplastic, immune-modulating, metabolism regulating, anti-depressant, neuroprotective and tissue protective effects. However, its poor solubility and poor absorption in the free form in the gastrointestinal tract and its rapid biotransformation to inactive metabolites greatly limit its utility as a health-promoting agent and dietary supplement. Recent advances in micro- and nano-formulations of curcumin with greatly enhanced absorption resulting in desirable blood levels of the active forms of curcumin now make it possible to address a wide range of potential applications, including pain management, and as tissue protective. Using these forms of highly bioavailable curcumin now enable a broad spectrum of appropriate studies to be conducted. This review discusses the formulations designed to enhance bioavailability, metabolism of curcumin, relationships between solubility and particle size relative to bioavailability, human pharmacokinetic studies involving formulated curcumin products, the widely used but inappropriate practice of hydrolyzing plasma samples for quantification of blood curcumin, current applications of curcumin and its metabolites and promising directions for health maintenance and applications.


Subject(s)
Blood Cells/chemistry , Curcumin/pharmacokinetics , Animals , Biological Availability , Curcumin/chemistry , Drug Compounding , Humans , Particle Size , Solubility , Theranostic Nanomedicine
5.
Phytother Res ; 34(8): 1838-1846, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32101364

ABSTRACT

Confusion and misunderstanding exist regarding the lack of cardiovascular and other adverse health effects of p-synephrine and p-octopamine relative to ephedrine and m-synephrine (phenylephrine) which are known for their effects on the cardiovascular system. These four molecules have some structural similarities. However, the structural and stereochemical differences of p-synephrine and p-octopamine as related to ephedrine and m-synephrine result in markedly different adrenergic receptor binding characteristics as well as other mechanistic differences which are reviewed. p-Synephrine and p-octopamine exhibit little binding to α-1, α-2, ß-1 and ß-2 adrenergic receptors, nor are they known to exhibit indirect actions leading to an increase in available levels of endogenous norepinephrine and epinephrine at commonly used doses. The relative absence of these mechanistic actions provides an explanation for their lack of production of cardiovascular effects at commonly used oral doses as compared to ephedrine and m-synephrine. As a consequence, the effects of ephedrine and m-synephrine cannot be directly extrapolated to p-synephrine and p-octopamine which exhibit significantly different pharmacokinetic, and physiological/pharmacological properties. These conclusions are supported by human, animal and in vitro studies that are discussed.


Subject(s)
Ephedrine/therapeutic use , Octopamine/therapeutic use , Synephrine/therapeutic use , Animals , Ephedrine/pharmacology , Humans , Octopamine/pharmacology , Rats , Synephrine/pharmacology
6.
J Diet Suppl ; 17(3): 355-363, 2020.
Article in English | MEDLINE | ID: mdl-30835576

ABSTRACT

p-Synephrine is the primary active ingredient in bitter orange (Citrus aurantium) extract and is present in other citrus species. This review summarizes all known case reports that have been published regarding adverse events associated with multi-ingredient dietary supplements containing bitter orange extract. A common characteristic of all the case studies was the assumption that if bitter orange extract is listed on the label of the product it is the most likely cause of any adverse effect, although in no case was the presence of p-synephrine determined or a direct link demonstrated. No case study reviewed the existing published literature, and all failed to note that numerous clinical studies have not demonstrated adverse effects at commonly used doses. Most studies did not indicate the composition of the product involved, and no study analyzed the product in question. In no case was a direct correlation between the event and p-synephrine made. Although p-synephrine and ephedrine have some structural similarity, the structural differences result in markedly different pharmacokinetic, physiological, and pharmacological effects, and thus the effects produced by ephedrine cannot be extrapolated to p-synephrine.


Subject(s)
Dietary Supplements/adverse effects , Drug-Related Side Effects and Adverse Reactions/etiology , Plant Extracts/adverse effects , Single-Case Studies as Topic , Citrus , Humans , Synephrine/adverse effects
7.
BMC Complement Altern Med ; 19(1): 293, 2019 Nov 04.
Article in English | MEDLINE | ID: mdl-31684927

ABSTRACT

BACKGROUND: Numerous health benefits have been demonstrated for curcumin which is extracted from turmeric (Curcuma longa L). However, due to its poor absorption in the free form in the gastrointestinal tract and rapid biotransformation, various formulations have been developed to enhance its bioavailability. Previous studies indicate that the free form of curcumin is more bioactive than its conjugated counterparts in target tissues. Most curcumin pharmacokinetics studies in humans designed to assess its absorption and bioavailability have measured and reported total (free plus conjugated) curcumin, but not free, bioactive curcumin in the plasma because enzymatic hydrolysis was employed prior to its extraction and analysis. Therefore, the bioavailability of free curcumin cannot be determined. METHODS: Eight human subjects (4 male, 4 female) consumed a single dose of 400 mg curcumin in an enhanced absorption formulation, and blood samples were collected over 6 h. Plasma was treated either with or without glucuronidase/sulfatase prior to extraction. Curcumin and its major metabolites were analyzed using HPLC-tandem mass spectrometry. In addition, the literature was searched for pharmacokinetic studies involving curcumin using PubMed and Google Scholar, and the reported bioavailability data were compared based on whether hydrolysis of plasma samples was used prior to sample analysis. RESULTS: Hydrolysis of blood plasma samples prior to extraction and reporting the results as "curcumin" obscures the amount of free, bioactive curcumin and total curcuminoids as compared to non-hydrolyzed samples. As a consequence, the data and biological effects reported by most pharmacokinetic studies are not a clear indication of enhanced plasma levels of free bioactive curcumin due to product formulations, leading to a misrepresentation of the results of the studies and the products when enzymatic hydrolysis is employed. CONCLUSIONS: When enzymatic hydrolysis is employed as is the case with most studies involving curcumin products, the amount of free bioactive curcumin is unknown and cannot be determined. Therefore, extreme caution is warranted in interpreting published analytical results from biological samples involving ingestion of curcumin-containing products. TRIAL REGISTRATION: ClinicalTrails.gov, trial identifying number NCT04103788 , September 24, 2019. Retrospectively registered.


Subject(s)
Curcumin/analysis , Glucuronidase/chemistry , Plasma/chemistry , Sulfatases/chemistry , Curcuma/chemistry , Curcumin/metabolism , Female , Humans , Hydrolysis , Male , Middle Aged , Prospective Studies
8.
J Alzheimers Dis ; 69(3): 631-649, 2019.
Article in English | MEDLINE | ID: mdl-31127770

ABSTRACT

Neurons consume the highest amount of oxygen, depend on oxidative metabolism for energy, and survive for the lifetime of an individual. Therefore, neurons are vulnerable to death caused by oxidative-stress, accumulation of damaged and dysfunctional proteins and organelles. There is an exponential increase in the number of patients diagnosed with neurodegenerative diseases such as Alzheimer's (AD) as the number of elderly increases exponentially. Development of AD pathology is a complex phenomenon characterized by neuronal death, accumulation of extracellular amyloid-ß plaques and neurofibrillary tangles, and most importantly loss of memory and cognition. These pathologies are most likely caused by mechanisms including oxidative stress, mitochondrial dysfunction/stress, accumulation of misfolded proteins, and defective organelles due to impaired proteasome and autophagy mechanisms. Currently, there are no effective treatments to halt the progression of this disease. In order to treat this complex disease with multiple biochemical pathways involved, a complex treatment regimen targeting different mechanisms should be investigated. Furthermore, as AD is a progressive disease-causing morbidity over many years, any chemo-modulator for treatment must be used over long period of time. Therefore, treatments must be safe and non-interfering with other processes. Ideally, a treatment like medicinal food or a supplement that can be taken regularly without any side effect capable of reducing oxidative stress, stabilizing mitochondria, activating autophagy or proteasome, and increasing energy levels of neurons would be the best solution. This review summarizes progress in research on different mechanisms of AD development and some of the potential therapeutic development strategies targeting the aforementioned pathologies.


Subject(s)
Alzheimer Disease/pathology , Signal Transduction/drug effects , Alzheimer Disease/drug therapy , Animals , Autophagy , Disease Progression , Humans , Oxidative Stress
9.
Toxicol Sci ; 170(1): 20-24, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30968141

ABSTRACT

The Society of Toxicology announces the development of a Learning Framework (https://www.toxicology.org/education/docs/SOT-Toxicology-Learning-Objectives.pdf) for undergraduate toxicology that will facilitate the development and sharing of evidence-based teaching materials for undergraduate toxicology educators throughout the world. This Learning Framework was modeled on the "Vision and Change Report" (www.visionandchange.org), an effort of the National Science Foundation and American Association for the Advancement of Science defining Core Concepts and Core Competencies to inform undergraduate biology course design. Vision and Change (V&C) has gained national acceptance, becoming a foundation for 14 upper-level courses designed by professional life science scientific societies. The undergraduate toxicology Learning Framework includes 5 Core Concepts aligned with V&C that encompass the discipline of toxicology: Evolution; Biological Information, Risk and Risk Management; Systems Toxicology; and Pathways and Transformations for Energy and Matter. Underpinning the Core Concepts are Level 2 Toxicology Concepts, which are broad disciplinary categories, Level 3 Learning Objectives, which address specific learning goals, and Level 4 Example Learning Objectives and Case Studies, which provide examples of how content might be taught. Syllabi from more than 20 undergraduate toxicology courses and several undergraduate toxicology textbooks were surveyed to determine toxicology-related Learning Objectives. From these, undergraduate educators can design courses tailored to their institutional needs by selecting a subset of Learning Objectives. Publication of a Learning Framework for toxicology will enable integration into other disciplines and facilitate the development and sharing of evidenced-based teaching materials for toxicology to educators in allied disciplines. Ultimately this will expand toxicology's impact to a broader audience.


Subject(s)
Education, Professional , Societies, Scientific , Teaching/organization & administration , Toxicology/education , Curriculum , Education, Professional/methods , Education, Professional/organization & administration , Learning , Models, Educational , United States
10.
Curr Pharm Des ; 24(43): 5181-5187, 2018.
Article in English | MEDLINE | ID: mdl-30657034

ABSTRACT

BACKGROUND: Gallic acid (GA-3,4,5-trihydroxybenzoic acid), a phenolic phytochemical, is a ubiquitous secondary metabolite found in most plants, with appreciable concentrations in grapes seed, rose flowers, sumac, oak and witch hazel. GA often results from the hydrolysis of terpenes and the polyphenol tannic acid. APPLICATIONS: It exhibits powerful antioxidant, anti-inflammatory, antimicrobial, and anti-cancer activities. Most intriguing benefit has been reported to be on the skin. Due to these beneficial properties, GA and its derivatives (e.g. lipid-soluble phenols such as synthetic gallic esters aka gallates) have been extensively used as an adjuvant in a number of therapeutic formulations, as a substitute of hydrocortisone in children with atopic dermatitis (AD) and other skin conditions (hyperpigmentation, wound healing), and as a cosmetic ingredient. GA has a USFDA GRAS status (generally recognized as safe), exhibiting fairly low systemic toxicity and associated mortality at acute doses in many experimental models. Despite anti-skin aging benefits obtained with relatively safe GA formulations, few cases of gallate-induced skin allergic have been reported in humans. Therefore, approaches to improve the bioavailability and biodegradability of this poor-water soluble and non-biodegradable phenolic compound are warranted. PURPOSE: This review has focused on the recently reported biological activities pertaining to the skin as well as the pharmacological properties of GA and its derivatives with special emphasis on its use in (nano-) cosmetic formulations. Since this is an evolving area of research, an adequate emphasis has been placed upon advantages and disadvantages of various nanoformulations.


Subject(s)
Cosmeceuticals/pharmacology , Cosmetics/pharmacology , Gallic Acid/pharmacology , Nanoparticles/chemistry , Skin/drug effects , Animals , Cosmeceuticals/chemistry , Cosmetics/chemistry , Gallic Acid/chemistry , Humans
11.
Arch Toxicol ; 92(2): 669-677, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28980048

ABSTRACT

Age-related motor deficits, such as loss of balance and coordination, are caused, in part, by loss of dopaminergic neurons. Oxidative stress is known to play a role in this neuronal loss. Resveratrol, a natural antioxidant with anticancer and anti-inflammatory potential, has been shown to protect dopaminergic-like cells (SH-SY5Y) against oxidative stress. However, the low bioavailability of resveratrol makes it worthwhile to explore newer compounds with similar properties. Piceid (RV8), an analog of resveratrol, has greater bioavailability than resveratrol, and our studies found that piceid (10, 20, 30 µM) protects SH-SY5Y cells against oxidative stress. Our investigations also found that the neuroprotection afforded by piceid was decreased when the MAP kinases, ERK1/2 and ERK5, were independently inhibited. Since oxidative stress is considered a master operator of apoptosis, our study also scrutinized dopamine-induced apoptosis and whether caspase-3/7 and Bcl-2 are involved, following piceid pretreatment followed by dopamine exposure. Our findings suggested that piceid pretreatment inhibited the dopamine-induced increase in caspase-3/7 activity and dopamine-induced loss of Bcl-2 expression. Overall, these findings suggest that the neuroprotective effects of piceid are mediated via the activation of ERK1/2, ERK5, and inhibition of apoptosis caused by oxidative stress.


Subject(s)
Dopaminergic Neurons/drug effects , Glucosides/pharmacology , Neuroprotective Agents/pharmacology , Resveratrol/analogs & derivatives , Stilbenes/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Dopamine/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
Curr Neurovasc Res ; 12(4): 363-74, 2015.
Article in English | MEDLINE | ID: mdl-26265154

ABSTRACT

Type I Diabetes is characterized by the presence of hyperglycemia due to insulin deficiency and consequent impaired hepatic glucose metabolism. During diabetes, the liver becomes the most important tissue for the regulation of serum glucose. However, elevated glucose causes continuous oxidative damage to the liver, reducing its capacity to ameliorate hyperglycemia, which contributes to macrovascular complications [1]. Numerous epidemiological studies have demonstrated that excess human consumption of diets rich in specific bioflavonoid phytochemicals attenuates the effects of diabetes. Thus, this study was designed to investigate whether a bioflavonoid mixture could : i) attenuate streptozotocin (STZ)-induced hyperglycemia, ii) potentiate antioxidant signaling in the liver, and iii) ameliorate the apoptotic signaling cascade in the liver of STZ-induced hyperglycemic mice. In order to examine our hypothesis, three well-investigated antioxidant phytochemicals, curcumin, hesperidin and rutin, were combined into a mixture (CHR) for this study. Diabetes was induced in 6-month-old female ICR mice by STZ (100 mg/kg, i.p.) administration, and CHR or vehicle control was orally administered (200 mg/kg per body weight of each ingredient) to the hyperglycemic mice (blood glucose levels > 250 mg/dl) for a period of 14 days. Administration of CHR to the hyperglycemic mice significantly reduced blood glucose levels, attenuated STZ-induced lipid peroxidation and total nitrate/nitrite levels, and significantly augmented the expression of superoxide dismutase and glutathione in the liver. STZ-induced hyperglycemia resulted in downregulation of antiapoptotic proteins Bcl-2 by 66% and Bcl-XL by 51%, and upregulation of the pro-apoptotic Bad (69%) with an increase in the ratio of cytosolic/mitochondrial cytochrome c by 81% in hepatic tissue. Administration of CHR significantly ameliorated apoptotic signaling in STZ-induced diabetic mice, significantly increasing Bad/Bcl-2 and Bad/Bcl-XL ratios to 410% and 244% respectively in the hyperglycemic group. This study demonstrated that a bioflavonoid mixture of curcumin, hesperidin and rutin (CHR) ameliorates hepatic oxidative stress caused by STZ-induced hyperglycemia, resulting in improved hepatic function and glucose regulation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Curcumin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Oxidative Stress/drug effects , Signal Transduction/drug effects , Analysis of Variance , Animals , Blood Glucose , Disease Models, Animal , Female , Flavonoids/metabolism , Hesperidin/therapeutic use , Liver/pathology , Mice, Inbred ICR , Nitrates/metabolism , Oxidation-Reduction , Rutin/therapeutic use , Superoxide Dismutase
13.
Adv Drug Deliv Rev ; 81: 62-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450260

ABSTRACT

Hepatocellular carcinoma (HCC) remains one of the most lethal forms of cancer in the world. In this arena, utilities of microRNA (miRNA) as a sophisticated tool, in therapeutics and diagnostics, remains a prime focus among the leading researchers. It is well known that viral hepatitis, chronic alcoholism and metabolic syndrome are the prime causes of HCCs. Nevertheless, HCCs are usually diagnosed at late stages by using current serum biomarkers. Most epidemiological studies have found the survival rate in HCC cases to be relatively low. Therefore, development of effective noninvasive biomarkers for early detention of HCC and new strategies for HCC treatment remains a high priority research area on the shoulders of liver oncologists. Accumulating evidence in cancer diagnostics and therapeutics indicates that miRNAs involve in HCC progression, which may serve as sensitive biomarkers for detecting carcinogenesis and monitoring therapies of HCC. This review focuses on the role of miRNAs in the diagnostics and therapeutics of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Animals , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/therapy , Disease Progression , Early Detection of Cancer , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/therapy , Neoplasm Staging , Survival Rate
14.
Phytother Res ; 27(8): 1107-14, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22976133

ABSTRACT

Extracts and powders of Cissus quadrangularis have been used for many years to promote bone and tissues healing, as an analgesic, to treat infections, as an anabolic, and to promote weight loss and weight management. This review summarizes the studies in animals, humans and in vitro systems that have been conducted to determine the efficacy and safety of various Cissus preparations. Animal and in vitro studies provide support for the use of Cissus in promoting bone fracture healing and as an anti-osteoporotic. Several human studies support the use of Cissus extracts in weight management. No studies have been conducted demonstrating that Cissus exhibits anabolic and body building activities. Based on studies to date, Cissus extracts appear to be exceedingly safe and free of adverse effects at the doses commonly used. A wide variety of chemical constituents have been isolated and identified from Cissus extracts, including steroids, flavonoids, stilbenes, iridoids, triterpenes and gallic acid derivatives. However, in few cases have specific physiological effects been related to identifiable constituents. Better standardization of extracts and more well-controlled human studies are required.


Subject(s)
Cissus/chemistry , Plant Extracts/adverse effects , Plant Extracts/therapeutic use , Analgesics/adverse effects , Analgesics/therapeutic use , Animals , Anti-Infective Agents/adverse effects , Anti-Infective Agents/therapeutic use , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/therapeutic use , Antioxidants/adverse effects , Antioxidants/therapeutic use , Cissus/adverse effects , Clinical Trials as Topic , Disease Models, Animal , Humans , Osteogenesis/drug effects , Weight Loss/drug effects
15.
Curr Neurovasc Res ; 9(4): 282-95, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22873725

ABSTRACT

Doxorubicin (DOX), a prominent anticancer agent has enjoyed considerable popularity in the last few decades because of its usefulness in the management of various forms of cancers, but its organotoxic potential (cardio-, hepatoand nephrotoxicity) has constrained on its clinical use. This study investigated whether DOX has the ability to cause nephrotoxicity in vivo and if so, whether it is linked to oxidative stress (OS). Another important goal was to describe whether expression of pro- and anti-apoptotic genes in kidneys was driven by OS. In order to explore DOX's nephrotoxic potential, male rats (Sprague Dawley; 500-520g; fed ad libitum) were administered i.p. with a single dose of DOX (12 mg/kg) on day one and sacrificed seven days later (day 8). Changes in serum chemistries (i.e., serum urea nitrogen, SUN, and creatinine) were determined immediately upon sacrifice, whereas kidney tissues were subjected to several sensitive biomarkers for OS, such as, lipid peroxidation, Superoxide dismutase (SOD) activity and chromatin fragmentation. The most important goal was to evaluate the select expression of Apaf-1, Caspase-3, Bad, Bax, Bcl-2, Bcl-xL, p53 and Mdm2 genes in order to understand the underlying link between extrinsic and intrinsic pathways of cell death. Data revealed that DOX-exposed animals showed significant nephrotoxicity as reflected in increased SUN (5.6-fold) and creatinine (2.65 fold) levels with considerably decreased body weight. Increases in kidney injury markers reflected parallel elevations in lipid peroxidation (1.7-fold) and genomic DNA fragmentation (2.9 fold) coupled with a proportionate reduction in total SOD activity suggesting DOX-assaulted kidneys encountered massive OS. Western blot showed very striking changes: i) substantial increases in the expression of pro-apoptotic APAF-1, Caspase-3, Bax and Bad proteins; ii) Reduction in the expression of anti-apoptotic Bcl-2 and Bcl-xL genes; iii) considerable increase in the expression of p53 and suppression of its regulator Mdm2. Serum chemistry and tissue biochemistry mirrored histopathology. In conclusion, this study for the first time may have shown a close link between mitochondrial perturbations and cell death regulating genes during DOXinduced nephrotoxicity, and described DOX's potential to inflict kidney injury in addition to other organs during chemotherapy in clinical setting.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Apoptosis/drug effects , Doxorubicin/toxicity , Oxidative Stress/drug effects , Signal Transduction/drug effects , Acute Kidney Injury/metabolism , Animals , Apoptosis/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Oxidative Stress/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics
16.
Mol Cell Biochem ; 365(1-2): 351-61, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22441882

ABSTRACT

Treatment during early tumor development has greater success because tissue growth remains largely confined to its original locus. At later stages, malignant cells migrate from their original location, invade surrounding normal areas, and can disseminate widely throughout the body. Remodeling of the extracellular matrix (ECM) serves as a key facilitator of this dissemination. Proteolytic enzymes including plasmin and matrix metalloproteinases (MMPs) play an integral role in degrading the surrounding ECM proteins and clearing a path for tumor cell migration. Specific MMPs are highly expressed late during malignant tumor invasion. It is not understood whether early changes in MMPs influence apoptotic and necrotic cell death, processes known to govern the early stages of carcinogenesis. Similarly, the interaction between MDM2 and p53 is tightly controlled by a complex array of post-translational modifications, which in turn dictates the stability and activity of both p53 and MDM2. The present studies examine the hypothesis that model hepatotoxin dimethylnitrosamine (DMN), which is also a model carcinogen, will induce the MMP family of proteins after administration in hepatotoxic doses. Doses of 25, 50, and 100 mg/kg DMN were administered i.p. to male C3H mice. Changes in parameters associated with apoptotic and necrotic cell death, DNA damage, cell proliferation, and extracellular proteinases were examined in liver at 24 h. Serum ALT activity, oxidative stress [malondialdehyde], and caspase-activated DNAse mediated DNA laddering increased in a dose-dependent manner, as did the level of MDM2 protein. MMP-9, -10 and -12 (gelatinase-B, stromelysin-2, macrophage elastase), and p53 protein levels increased following 25 mg/kg DMN, but were successively decreased after higher DMN doses. The results of this study demonstrate changes in MDM2 and MMPs during DMN-induced acute liver injury and provide a plausible linkage between DMN-induced oxidative stress-mediated genomic injury and its likely involvement in setting the stage for initiating subsequent metastatic disease at later circumstances.


Subject(s)
Carcinogens/toxicity , DNA Fragmentation , Dimethylnitrosamine/toxicity , Liver/enzymology , Matrix Metalloproteinases, Secreted/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Gene Expression/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Matrix Metalloproteinases, Secreted/genetics , Mice , Mice, Inbred C3H , Necrosis/pathology , Oxidative Stress , Proto-Oncogene Proteins c-mdm2/genetics , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Suppressor Protein p53/genetics
17.
Curr Neurovasc Res ; 9(1): 58-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22272768

ABSTRACT

Curcumin (CUR; diferuloylmethane), a rhizome extract of Curcuma Longa L. is commonly used as a food coloring and flavoring agent. Although oriental and Ayurvedic medicines have traditionally used CUR in the treatment of diseases, conventional medicine has just begun to recognize its potential therapeutic value. Numerous recent studies have demonstrated the ability of CUR to halt or prevent certain types of cancer, decrease inflammation, and improve cardiovascular health. However, very few studies have examined its ability to protect against drug-induced organ injury. This study explored whether CUR pre-exposure has the potential to prevent acetaminophen (APAP)-induced: (i) hepatotoxicity, (ii) genomic injury, (iii) oxidative stress in the liver, and (iv) apoptotic and necrotic cell deaths in the liver in vivo. Additional goals were to investigate the interplay of pro- and anti-apoptotic genes and their ultimate impact on various forms of cell death. In order to study the CUR-APAP interaction, male B6C3F1 mice were gavaged with CUR (17 mg/kg/day, p.o.) for 12 days followed by a single APAP exposure (400 mg/kg, ip). Four groups of animals (control, CUR, APAP, CUR+APAP) were sacrificed 24 h after APAP exposure. The results indicated that APAP-induced liver injury associated events as serum ALT (80-fold), lipid peroxidation (357%) and DNA fragmentation (469%) were markedly reduced to 3-fold, 134% and 162%, respectively, in the CUR+APAP group. The APAP-induced increase in expression of pro-apoptotic genes (Bax, caspase-3) decreased while expression of anti-apoptotic genes (Bcl-XL) increased in CUR preexposed mouse livers, and these changes were mirrored in the pattern of apoptotic and necrotic cell deaths. Levels of DNA damage sensor P5³ and its counterpart Mdm2 were also analyzed during this interaction. Based on the available literature, and these results, it seems likely that CUR may impart global protection in vivo against drug-induced liver injury by opposing several crucial events instrumental to both apoptosis and necrosis.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Curcumin/pharmacology , Signal Transduction/drug effects , Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Animals , Blotting, Western , DNA Fragmentation/drug effects , Gene Expression/drug effects , Lipid Peroxidation/drug effects , Male , Mice , Oxidative Stress/drug effects
18.
J Toxicol ; 2011: 683728, 2011.
Article in English | MEDLINE | ID: mdl-21785590

ABSTRACT

Besides fluorine, oxygen is the most electronegative element with the highest reduction potential in biological systems. Metabolic pathways in mammalian cells utilize oxygen as the ultimate oxidizing agent to harvest free energy. They are very efficient, but not without risk of generating various oxygen radicals. These cells have good antioxidative defense mechanisms to neutralize these radicals and prevent oxidative stress. However, increased oxidative stress results in oxidative modifications in lipid, protein, and nucleic acids, leading to mitochondrial dysfunction and cell death. Oxidative stress and mitochondrial dysfunction have been implicated in many neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, and stroke-related brain damage. Research has indicated mitochondria play a central role in cell suicide. An increase in oxidative stress causes mitochondrial dysfunction, leading to more production of reactive oxygen species and eventually mitochondrial membrane permeabilization. Once the mitochondria are destabilized, cells are destined to commit suicide. Therefore, antioxidative agents alone are not sufficient to protect neuronal loss in many neurodegenerative diseases. Combinatorial treatment with antioxidative agents could stabilize mitochondria and may be the most suitable strategy to prevent neuronal loss. This review discusses recent work related to oxidative toxicity in the central nervous system and strategies to treat neurodegenerative diseases.

19.
Oxid Med Cell Longev ; 3(2): 129-44, 2010.
Article in English | MEDLINE | ID: mdl-20716937

ABSTRACT

A novel dietary supplement composed of three well-known phytochemicals, namely, Salvia officinalis (sage) extract, Camellia sinensis (oolong tea) extract, and Paullinia cupana (guarana) extract, and two prominent vitamins (thiamine and niacin) was designed to provide nutritional support by enhancing metabolism and maintaining healthy weight and energy. The present study evaluated the safety of this dietary supplement (STG; S=sage; T=tea; G=guarana) and assessed changes in target organ antioxidant enzymes (liver, kidneys and heart), serum chemistry profiles and organ histopathology in Fisher 344 rats. Adult male and female Fisher 344 rats were fed control (no STG) or STG containing (1X and 7X, 1X=daily human dose) diets and sacrificed after 2 and 4 months. Serum chemistry analysis and histopathological examination of three vital target organs disclosed no adverse influence on protein, lipid and carbohydrate profiles, genomic integrity of the liver and/or the tissue architecture. However, analysis of the most important antioxidant components in the liver, kidney and heart homogenates revealed a dramatic increase in total glutathione concentrations, glutathione peroxidase and superoxide dismutase enzyme activities. Concomitantly, oxidative stress levels (malondialdehyde accumulation) in these three organs were less than control. Organ specific serum markers (ALT/AST for the liver; CPK/AST for the heart; BUN/creatinine for kidneys) and the genomic integrity disclosed no STG-induced alteration. Some of the serum components (lipid and protein) showed insignificant changes. Overall, STG-exposed rats were more active, and the results suggest that STG exposure produces normal serum chemistry coupled with elevated antioxidant capacity in rats fed up to seven times the normal human dose and does not adversely influence any of the vital target organs. Additionally, this study reiterates the potential benefits of exposure to a pharmacologically relevant combination of phytochemicals compared to a single phytochemical entity.


Subject(s)
Dietary Supplements , Heart/drug effects , Kidney/enzymology , Liver/enzymology , Myocardium/enzymology , Plant Extracts/pharmacology , Vitamins/pharmacology , Animals , Antioxidants/metabolism , Camellia sinensis/chemistry , Enzyme Activation/drug effects , Female , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Kidney/drug effects , Liver/drug effects , Male , Niacin/pharmacology , Paullinia/chemistry , Rats , Rats, Inbred F344 , Salvia officinalis/chemistry , Superoxide Dismutase/metabolism , Thiamine/pharmacology , Vitamins/chemistry
20.
Toxicol Appl Pharmacol ; 245(2): 143-52, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20144634

ABSTRACT

The emergence of silymarin (SMN) as a natural remedy for liver diseases, coupled with its entry into NIH clinical trial, signifies its hepatoprotective potential. SMN is noted for its ability to interfere with apoptotic signaling while acting as an antioxidant. This in vivo study was designed to explore the hepatotoxic potential of Doxorubicin (Dox), the well-known cardiotoxin, and in particular whether pre-exposures to SMN can prevent hepatotoxicity by reducing Dox-induced free radical mediated oxidative stress, by modulating expression of apoptotic signaling proteins like Bcl-xL, and by minimizing liver cell death occurring by apoptosis or necrosis. Groups of male ICR mice included Control, Dox alone, SMN alone, and Dox with SMN pre/co-treatment. Control and Dox groups received saline i.p. for 14 days. SMN was administered p.o. for 14 days at 16 mg/kg/day. An approximate LD(50) dose of Dox, 60 mg/kg, was administered i.p. on day 12 to animals receiving saline or SMN. Animals were euthanized 48 h later. Dox alone induced frank liver injury (>50-fold increase in serum ALT) and oxidative stress (>20-fold increase in malondialdehyde [MDA]), as well as direct damage to DNA (>15-fold increase in DNA fragmentation). Coincident genomic damage and oxidative stress influenced genomic stability, reflected in increased PARP activity and p53 expression. Decreases in Bcl-xL protein coupled with enhanced accumulation of cytochrome c in the cytosol accompanied elevated indexes of apoptotic and necrotic cell death. Significantly, SMN exposure reduced Dox hepatotoxicity and associated apoptotic and necrotic cell death. The effects of SMN on Dox were broad, including the ability to modulate changes in both Bcl-xL and p53 expression. In animals treated with SMN, tissue Bcl-xL expression exceeded control values after Dox treatment. Taken together, these results demonstrated that SMN (i) reduced, delayed onset, or prevented toxic effects of Dox which are typically associated with hydroxyl radical production, (ii) performed as an antioxidant limiting oxidative stress, (iii) protected the integrity of the genome, and (iv) antagonized apoptotic and necrotic cell death while increasing antiapoptotic Bcl-xL protein levels and minimizing the leakage of proapoptotic cytochrome c from liver mitochondria. These observations demonstrate the protective actions of SMN in liver, and raise the possibility that such protection may extend to other organs during Dox treatment including the heart.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Doxorubicin/toxicity , Oxidative Stress/drug effects , Silymarin/pharmacology , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Deoxyribonucleases/metabolism , Male , Mice , Mice, Inbred ICR , Mitochondria, Liver/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL