Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Saudi J Biol Sci ; 29(5): 3414-3424, 2022 05.
Article in English | MEDLINE | ID: mdl-35844406

ABSTRACT

Lung injuries are attributed due to exposure to Drugs or chemicals. One of the important challenging situations for the clinicians is to manage treatments of different diseases with acute lung injury (ALI). The objective of this study was to investigate the possible protective mechanisms and action of a novel Phosphodiesterase-4 inhibitor "Apremilast" (AP) in lipopolysaccharide (LPS)-induced lung injury. Blood sample from each animals were collected in a vacuum blood collection tube. The rat lungs were isolated for oxidative stress assessment, western blot analysis and their mRNA expressions using RT-PCR. Exposure of LPS in rats causes significant increase in oxidative stress, activates the pro-inflammatory cytokines release like tissue necrotic factor-alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6), modulated gene expression, protein expression and histopathological changes which were reversed by administration of AP. Finding of the research enlighten the protective role of AP against LPS-induced ALI.

3.
Cardiovasc Toxicol ; 19(4): 344-356, 2019 08.
Article in English | MEDLINE | ID: mdl-30644033

ABSTRACT

This study has been initiated to investigate whether sunitinib (SUN) alters the expression of key genes engaged in mitochondrial transport and oxidation of long chain fatty acids (LCFA), and if so, whether these alterations should be viewed as a mechanism of SUN-induced cardiotoxicity, and to explore the molecular mechanisms whereby carnitine supplementation could attenuate SUN-induced cardiotoxicity. Adult male Wister albino rats were assigned to one of the four treatment groups: Rats in group 1 received no treatment but free access to tap water for 28 days. Rats in group 2 received L-carnitine (200 mg/kg/day) in drinking water for 28 days. Rats in group 3 received SUN (25 mg/kg/day) in drinking water for 28 days. Rats in group 4 received the same doses of L-carnitine and SUN in drinking water for 28 days. Treatment with SUN significantly increased heart weight, cardiac index, and cardiotoxicity enzymatic indices, as well as severe histopathological changes. Moreover, SUN significantly decreased level of adenosine monophosphate-activated protein kinase (AMPKα2), total carnitine, adenosine triphosphate (ATP) and carnitine palmitoyltransferase I (CPT I) expression and significantly increased acetyl-CoA carboxylase-2 (ACC2) expression and malonyl-CoA level in cardiac tissues. Interestingly, carnitine supplementation resulted in a complete reversal of all the biochemical, gene expression and histopathological changes-induced by SUN to the control values. In conclusion, data from this study suggest that SUN inhibits AMPK downstream signaling with the consequent inhibition of mitochondrial transport of LCFA and energy production in cardiac tissues. Carnitine supplementation attenuates SUN-induced cardiotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/toxicity , Carnitine/pharmacology , Dietary Supplements , Energy Metabolism/drug effects , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Sunitinib/toxicity , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiotoxicity , Carnitine O-Palmitoyltransferase/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Male , Malonyl Coenzyme A/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Rats, Wistar , Signal Transduction
4.
Inflammopharmacology ; 27(4): 817-827, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30600471

ABSTRACT

BACKGROUND: Carfilzomib (CFZ), a proteasome inhibitor approved by the FDA to treat multiple myeloma, may cause nephrotoxicity. HYPOTHESIS: Rutin is a bioflavonoid with antioxidant properties. We aimed to examine whether rutin protects the kidney from CFZ-induced nephrotoxicity. STUDY DESIGN: This study aimed to demonstrate the effect of rutin on CFZ-induced renal injury via the inhibition of oxidative stress and inflammation. METHODS: Wistar albino rats were divided into six groups (n = 6): Group 1 (normal control; NC) was administered normal saline for 3 weeks; Group 2 (CFZ/toxic group) received CFZ [4 mg/kg, intraperitoneal (i.p.) injection] twice weekly for 3 weeks; Group 3 (standard treatment group) was administered CFZ (4 mg/kg, i.p.) and olmesartan (2 mg/kg, p.o.) for 3 weeks; Group 4 was administered CFZ (4 mg/kg, i.p.) and rutin (10 mg/kg, p.o.) for 3 weeks; Group 5 was administered CFZ (4 mg/kg, i.p.) and rutin (20 mg/kg, p.o.) for 3 weeks; and Group 6 was administered CFZ (4 mg/kg, i.p.) and rutin (40 mg/kg, p.o.) for 3 weeks. We carried out haematological and biochemical analyses, determined oxidative stress, caspase-3 activity, and protein levels, and performed a histopathological evaluation to confirm CFZ-induced nephrotoxicity and its prevention by rutin administration. RESULTS: Exposure to only CFZ significantly (p < 0.05) increased white blood cell (WBC) count, Hb%, and HTC% concentration; however, these features were significantly decreased (p < 0.05) when olmesartan and rutin were administered. CFZ administration significantly decreased (p < 0.0001) the level of antioxidant enzymes; whereas, administration of olmesartan and rutin significantly reversed (p < 0.05) their levels toward the normal range. The levels of caspase-3 enzyme significantly increased (p < 0.001) in the CFZ group and were reduced toward the normal values by olmesartan and rutin administration. Furthermore, the results of NOS-2, NF-κB, IkBa, and IL-17 protein estimation and the histopathological evaluation strengthened our findings that rutin exhibits a protective effect against CFZ-induced nephrotoxicity. CONCLUSION: These findings clearly demonstrate that rutin ameliorates CFZ-induced oxidative stress and inflammation in nephrotoxicity via the NOS-mediated NF-κB signaling pathway.


Subject(s)
Inflammation/drug therapy , Nitric Oxide Synthase/metabolism , Oligopeptides/pharmacology , Oxidative Stress/drug effects , Rutin/pharmacology , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Flavonoids/pharmacology , Imidazoles/pharmacology , Inflammation/metabolism , Kidney/drug effects , Kidney/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Wistar , Tetrazoles/pharmacology
5.
Asian Pac J Cancer Prev ; 19(3): 777-783, 2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29582634

ABSTRACT

Background: Breast cancer is affected by the immune system in that different cytokines play roles in its initiation and progression. Interleukin-10 (IL-10), an anti-inflammatory cytokine, is an immunosuppressive factor involved in tumorigenesis. The present study was conducted to investigate the gene silencing effect of a small interference RNA (siRNA) targeting IL-10 on the apoptotic pathway in breast cancer cell line. Methods: The siRNA targeting IL-10 and a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) clone were introduced into MDA-MB-231 cells. Real-time PCR assays were used to determine IL-10 and GAPDH gene expression levels, in addition to those for protein kinase B (AKT), phosphoinositide 3-kinase (PI3K), B-cell lymphoma 2 (Bcl2), caspase-3 and caspase-9 genes related to apoptosis. Results: Inhibition of IL-10 by the siRNA accelerated apoptosis and was accompanied by significant increase in caspase-3 and caspase-9 and a significant decrease in PI3K, AKT and Bcl2 expression levels compared to the non-transfected case. Conclusions: In conclusion, the production of IL-10 may represent a new escape mechanism by breast cancer cells to evade destruction by the immune system. IL-10 gene silencing causes down regulation of both PI3K/AKT and Bcl2 gene expression and also increases the Bbc3, BAX caspase3, and caspase 3 cleavage expression levels. IL­10 might represent a promising new target for therapeutic strategies.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Silencing , Interleukin-10/antagonists & inhibitors , RNA, Small Interfering/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Signal Transduction , Tumor Cells, Cultured
6.
BMC Complement Altern Med ; 17(1): 472, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28962559

ABSTRACT

BACKGROUND: Cisplatin is widely used chemotherapeutic agent for cancer treatment with limited uses due to its neurotoxic side effect. The aim of this study was to determine the potential preventive effects of rutin on the brain of cisplatin- neurotoxic rat model. METHODS: Forty rats were divided into four groups. Group-1 (control group) was intra-peritoneal (IP) injected with 2.5 ml/kg saline. Group-2 (rutin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days. Group-3 (cisplatin group) was IP received 5 mg/kg cisplatin single dose. Group-4 (rutin and cisplatin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days with a single dose of 5 mg/kg cisplatin IP on day ten. Brain tissues from frontal cortex was used to extract RNA, the gene expression levels of paraoxonase-1 (PON-1), PON-2, PON-3, peroxisome proliferator-activated receptor delta (PPAR-δ), and glutathione peroxidase (GPx) was investigated by Real-time PCR. RESULTS: Cisplatin significantly decreased the expression levels of PON-1, PON-3, PPAR-δ and GPX whereas significantly increased PON-2 expression levels. Co-administration of Rutin prevented the cisplatin-induced toxicity by restoring the alteration in the studied genes to normal values as in the control group. CONCLUSION: This study showed that Rutin has neuroprotective effect and reduces cisplatin- neurotoxicity with possible mechanism via the antioxidant pathway.


Subject(s)
Brain/drug effects , Cisplatin/adverse effects , Neuroprotective Agents/pharmacology , Rutin/pharmacology , Animals , Body Weight/drug effects , Brain/enzymology , Brain/metabolism , Brain Chemistry/drug effects , Glutathione/metabolism , Glutathione Peroxidase/analysis , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Male , Oxidative Stress/drug effects , PPAR delta/analysis , PPAR delta/genetics , PPAR delta/metabolism , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
7.
BMC Nephrol ; 18(1): 194, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28619064

ABSTRACT

BACKGROUND: Cisplatin (CP) is commonly used in the treatment of different types of cancer but nephrotoxicity has been a major limiting factor. Therefore, the present study aimed to study the possible protective effect of rutin against nephrotoxicity induced by cisplatin in rats. METHODS: Forty male Wistar albino rats were randomly divided into 4 groups. Rats of group 1 control group intraperitoneal (i.p.) received 2.5 ml/kg, group 2 CP group received single dose 5 mg/kg cisplatin i.p. group 3 rutin group orally received 30 mg/kg rutin group 4 (CP plus rutin) received CP and rutin as in group 2 and 3. Kidneys were harvested for histopathology and for the study the gene expression of c-Jun N-terminal kinases (JNK), Mitogen-activated protein kinase 4 (MKK4), MKK7, P38 mitogen-activated protein kinases (P38), tumor necrosis factors alpha (TNF-α), TNF Receptor-Associated Factor 2 (TRAF2), and interleukin-1 alpha (IL-1-α). RESULTS: The cisplatin single dose administration to rats induced nephrotoxicity associated with a significant increase in blood urea nitrogen (BUN) and serum creatinine and significantly increase Malondialdehyde (MDA) in kidney tissues by 230 ± 5.5 nmol/g compared to control group. The animal treated with cisplatin showed a significant increase in the expression levels of the IL-1α (260%), TRFA2 (491%), P38 (410%), MKK4 (263%), MKK7 (412%), JNK (680%) and TNF-α (300%) genes compared to control group. Additionally, histopathological examination showed that cisplatin-induced interstitial congestion, focal mononuclear cell inflammatory, cell infiltrate, acute tubular injury with reactive atypia and apoptotic cells. Rutin administration attenuated cisplatin-induced alteration in gene expression and structural and functional changes in the kidney. Additionally, histopathological examination of kidney tissues confirmed gene expression data. CONCLUSION: The present study suggested that the anti-oxidant and anti-inflammatory effect of rutin may prevent CP-induced nephrotoxicity via decreasing the oxidative stress, inhibiting the interconnected ROS/JNK/TNF/P38 MAPK signaling pathways, and repairing the histopathological changes against cisplatin administration.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Acute Kidney Injury/pathology , Animals , Male , Rats , Rats, Wistar , Rutin , Treatment Outcome
8.
Mol Cell Neurosci ; 82: 76-87, 2017 07.
Article in English | MEDLINE | ID: mdl-28465254

ABSTRACT

Autism spectrum disorder (ASD) is neurodevelopmental disorders characterized by stereotypical repetitive behavior, impaired social interaction, and deficits in communication. The BTBR T+ Itpr3tf/J (BTBR) mice have been extensively used as an animal model of the ASD-like phenotype. Adenosine A2A receptors (A2ARs) are considered potential targets in the treatment of neurodegenerative diseases. In this study, we used the A2AR antagonist SCH 5826 (SCH) and the A2AR agonist CGS 21680 (CGS) to investigate the activation of A2AR signaling in immune cells. Further, we examined the effects of A2ARs on the expression of the cytokines interleukin 2 (IL-2), IL-6, IL-9, interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and transforming growth factor ß (TGF-ß) in the spleen and in splenic CD4+ T cells. In addition, we assessed the mRNA and protein expression levels of these cytokines in the brain tissue. Our results showed that the levels of IL-2+, IL-6+, IL-9+, IFN-γ+, and TNF-α+ were significantly lower, whereas the levels of TGF-ß+ in the spleen and in splenic CD4+ T cells were significantly higher in the CGS-treated mice than in the BTBR control and SCH-treated mice. In addition, reverse transcription polymerase chain reaction (RT-PCR) and western blot analysis showed a decrease in the mRNA and protein expression levels of IL-2, IL-6, IL-9, IFN-γ+, and TNF-α+ and an increase in the mRNA and protein expression levels of TGF-ß in the CGS-treated mice, while treatment with BTBR alone and SCH resulted in increased Th1 levels and decreased Th2 levels in the brain tissue. Our results suggest that treatment the A2AR agonist CGS may be a promising therapeutic option for neuroimmune dysfunction.


Subject(s)
Brain/metabolism , Cytokines/metabolism , Receptor, Adenosine A2A/drug effects , Signal Transduction , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Brain/drug effects , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenethylamines/pharmacology , Signal Transduction/drug effects
9.
BMC Complement Altern Med ; 17(1): 45, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28086769

ABSTRACT

BACKGROUND: Liver diseases are major global health problems. Ginseng extract has antioxidant, immune-modulatory and anti-inflammatory activities. This study investigated the effect of ginseng extract on carbon tetrachloride (CCl4)-induced liver fibrosis in rats. METHODS: Male Wistar rats were divided into four groups: control group, ginseng group, CCl4 group and CCl4 + ginseng group. Liver injury was induced by the intraperitoneal (I.P) injection of 3 ml/kg CCl4 (30% in olive oil) weekly for 8 weeks. The control group was I.P injected with olive oil. The expression of genes encoding transforming growth factor beta (TGF-ß), type I TGF-ß receptor (TßR-1), type II TGF-ß receptor (TßR-II), mothers against decapentaplegic homolog 2 (Smad2), Smad3, Smad4, matrix metalloproteinase 2 (MMP2), MMP9, tissue inhibitor matrix metalloproteinase-1 (TIMP-1), Collagen 1a2 (Col1a2), Collagen 3a1 (Col3a1), interleukin-8 (IL-8) and interleukin -10 (IL-10) were measured by real-time PCR. RESULTS: Treatment with ginseng extract decreased hepatic fat deposition and lowered hepatic reticular fiber accumulation compared with the CCl4 group. The CCl4 group showed a significant increase in hepatotoxicity biomarkers and up-regulation of the expression of genes encoding TGF-ß, TßR-I, TßR-II, MMP2, MMP9, Smad-2,-3, -4, and IL-8 compared with the control group. However, CCl4 administration resulted in the significant down-regulation of IL-10 mRNA expression compared with the control group. Interestingly, ginseng extract supplementation completely reversed the biochemical markers of hepatotoxicity and the gene expression alterations induced by CCl4. CONCLUSION: ginseng extract had an anti-fibrosis effect via the regulation of the TGF-ß1/Smad signaling pathway in the CCl4-induced liver fibrosis model. The major target was the inhibition of the expression of TGF-ß1, Smad2, and Smad3.


Subject(s)
Liver Cirrhosis/drug therapy , Panax/chemistry , Plant Extracts/administration & dosage , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride/adverse effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/genetics
10.
Mol Neurobiol ; 54(6): 4390-4400, 2017 08.
Article in English | MEDLINE | ID: mdl-27344332

ABSTRACT

Autism is a neurodevelopmental disorder characterized by stereotypic repetitive behaviors, impaired social interactions, and communication deficits. Numerous immune system abnormalities have been described in individuals with autism including abnormalities in the ratio of Th1/Th2/Th17 cells; however, the expression of the transcription factors responsible for the regulation and differentiation of Th1/Th2/Th17/Treg cells has not previously been evaluated. Peripheral blood mononuclear cells (PBMCs) from children with autism (AU) or typically developing (TD) control children were stimulated with phorbol-12-myristate 13-acetate (PMA) and ionomycin in the presence of brefeldin A. The expressions of Foxp3, RORγt, STAT-3, T-bet, and GATA-3 mRNAs and proteins were then assessed. Our study shows that children with AU displayed altered immune profiles and function, characterized by a systemic deficit of Foxp3+ T regulatory (Treg) cells and increased RORγt+, T-bet+, GATA-3+, and production by CD4+ T cells as compared to TD. This was confirmed by real-time PCR (RT-PCR) and western blot analyses. Our results suggest that autism impacts transcription factor signaling, which results in an immunological imbalance. Therefore, the restoration of transcription factor signaling may have a great therapeutic potential in the treatment of autistic disorders.


Subject(s)
Autistic Disorder/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Transcription Factors/metabolism , Autistic Disorder/genetics , Child , Child, Preschool , Flow Cytometry , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics
11.
Biochem Pharmacol ; 124: 69-82, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27984001

ABSTRACT

Psoriasis is an autoimmune inflammatory skin disease characterized by activated IL-23/STAT3/Th17 axis. Recently psoriatic inflammation has been shown to be associated with asthma. However, no study has previously explored how psoriatic inflammation affects airway inflammation. Therefore, this study investigated the effect of imiquimod (IMQ)-induced psoriatic inflammation on cockroach extract (CE)-induced airway inflammation in murine models. Mice were subjected to topical and intranasal administration of IMQ and CE to develop psoriatic and airway inflammation respectively. Various analyses in lung/spleen related to inflammation, Th17/Th2/Th1 cell immune responses, and their signature cytokines/transcription factors were carried out. Psoriatic inflammation in allergic mice was associated with increased airway inflammation with concurrent increase in Th2/Th17 cells/signature cytokines/transcription factors. Splenic CD4+ T and CD11c+ dendritic cells in psoriatic mice had increased STAT3/RORC and IL-23 mRNA expression respectively. This led us to explore the effect of systemic IL-23/STAT3 signaling on airway inflammation. Topical application of STA-21, a small molecule STAT3 inhibitor significantly reduced airway inflammation in allergic mice having psoriatic inflammation. On the other hand, adoptive transfer of IL-23-treated splenic CD4+ T cells from allergic mice into naive recipient mice produced mixed neutrophilic/eosinophilic airway inflammation similar to allergic mice with psoriatic inflammation. Our data suggest that systemic IL-23/STAT3 axis is responsible for enhanced airway inflammation during psoriasis. The current study also suggests that only anti-asthma therapy may not be sufficient to alleviate airway inflammatory burden in asthmatics with psoriasis.


Subject(s)
Bronchitis/complications , Disease Models, Animal , Hypersensitivity/complications , Interleukin-23/metabolism , Psoriasis/complications , STAT3 Transcription Factor/metabolism , Signal Transduction , Allergens/administration & dosage , Animals , Bronchoalveolar Lavage Fluid , CD4-Positive T-Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C
12.
Immunobiology ; 222(2): 206-217, 2017 02.
Article in English | MEDLINE | ID: mdl-27717524

ABSTRACT

We set out to investigate the influence of STA-21, a dynamic STAT-3 inhibitor, on the expansion and progression of rheumatoid arthritis (RA), and to determine its potential mechanisms of action in a mouse model of collagen antibody-induced arthritis (CAIA). To this end, arthritis was induced via intravenous (IV) injection of Balb/c mice with a cocktail of antibodies directed against type II collagen (1.5µg/mouse, IV), followed by lipopolysaccharide (LPS) at a dose of (25µg/mouse, i.p.) on day 3. Mice were then left untreated or were simultaneously treated with STA-21 (0.5mg/kg, i.p., once daily for 2 weeks) followed by evaluation for clinical and histological features of arthritic inflammation and flow cytometric analysis of cytokines and transcription factors in peripheral blood. STA-21 enhanced the clinical course of arthritis in CAIA mice and decreased CD8+RORγt+ and CD8+IL-21+ cells while inducing the production of CD8+Foxp3+ cells. Furthermore, STA-21 prevented the production of TNF-α and IL-6 in peripheral blood and increased IL-27 production by CD14+ cells. Moreover, STA-21 not only regulates Th1/Th2 serum cytokine levels but also the mRNA and protein expression of key factors including NF-κB p65, RORγt, T-bet, IL-4, GATA-3, JAK1, Stat3, and IL-21. Thus, administration of the Stat3 inhibitor STA-21 inhibits cellular signaling pathways and downstream activation of key transcription factors previously shown to play key roles in the pathogenesis of RA. Therefore, these data suggest that STA-21 could be considered as a potential treatment for patients with RA.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Polycyclic Compounds/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Asian Pac J Cancer Prev ; 17(11): 4965-4971, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28032724

ABSTRACT

Objective: Breast cancer is global female health problem worldwide. Most of the currently used agents for breast cancer treatment have toxic side-effects. Ginseng root, an oriental medicine, has many health benefits and may exhibit direct anti-cancer properties. This study was performed to assess the effects of ginseng on breast cancer cell lines. Materials and Methods: Cytotoxicity of ginseng extract was measured by MTT assay after exposure of MDA-MB-231, MCF-10A and MCF-7 breast cancer cells to concentrations of 0.25, 0.5, 1, 1.5, 2 and 2.5 mg/well. Expression levels of p21WAF, p16INK4A, Bcl-2, Bax and P53 genes were analyzed by quantitative real time PCR. Results: The treatment resulted in inhibition of cell proliferation in a dose-and time-dependent manner. p53, p21WAF1and p16INK4A expression levels were up-regulated in ginseng treated MDA-MB-231 and MCF-7 cancer cells compared to untreated controls and in MCF-10A cells. The expression levels of Bcl2 in the MDA-MB-231 and MCF-7 cells were down-regulated. In contrast, that of Bax was significantly up-regulated. Conclusion: The results of this study revealed that ginseng may inhibit breast cancer cell growth by activation of the apoptotic pathway.

14.
15.
PLoS One ; 11(9): e0163703, 2016.
Article in English | MEDLINE | ID: mdl-27690233

ABSTRACT

Dexrazoxane has been approved to treat anthracycline-induced cardiomyopathy and extravasation. However, the effect of dexrazoxane on epirubicin-induced genetic alterations in germ cells has not yet been reported. Thus, the aim of this study was to determine whether dexrazoxane modulates epirubicin-induced genetic damage in the germ cells of male mice. Our results show that dexrazoxane was not genotoxic at the tested doses. Furthermore, it protected mouse germ cells against epirubicin-induced genetic alterations as detected by the reduction in disomic and diploid sperm, spermatogonial chromosomal aberrations, and abnormal sperm heads. The attenuating effect of dexrazoxane was greater at higher dose, indicating a dose-dependent effect. Moreover, sperm motility and count were ameliorated by dexrazoxane pretreatment. Epirubicin induced marked biochemical changes characteristic of oxidative DNA damage including elevated 8-hydroxy-2'-deoxyguanosine levels and reduction in reduced glutathione. Pretreatment of mice with dexrazoxane before epirubicin challenge restored these altered endpoints. We conclude that dexrazoxane may efficiently mitigate the epirubicin insult in male germ cells, and prevent the enhanced risk of abnormal reproductive outcomes and associated health risks. Thus, pretreating patients with dexrazoxane prior to epirubicin may efficiently preserve not only sperm quality but also prevent the transmission of genetic damage to future generations.

16.
Mol Immunol ; 78: 65-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27608299

ABSTRACT

Protein tyrosine kinases are key mediators of the signal transduction cascades that control expression of many genes involved in the induction of inflammation caused by arthritis. Here we investigate the effect of the tyrosine kinase inhibitor tyrphostin AG126 on a mouse model of adjuvant-induced arthritis (AIA). We report that when given at 5mg/kg i.p. every 48h from days 0-21, AG126 exerts potent anti-arthritic effects. Further, we investigated the role of AG126 on the key mediators of arthritic inflammation, namely, edema, arthritic score, presence of immunophenotypes including Foxp3+, CD4+Foxp3+, and CD25+Foxp3+ T regulatory (Treg) cells, as well as pro- and anti-inflammatory mediators. AG126 treatment significantly attenuated the severity of AIA and caused a substantial reduction in the percentage of CD2+, CD3+, CD4+, CD8+, CD23+, CD80+, CD86+ CD122+, CD195+, TCRß+, and GITR+ cells in whole blood. Moreover, administration of AG126 under arthritis-inducing conditions resulted in suppression of IL-17A+, IFN-γ+, CD4+ and CD25+ populations while causing an increase in the Foxp3+, CD4+Foxp3+, and CD25+Foxp3+ Treg populations in the spleen. In addition, RT-PCR analysis revealed increased expression of CD4, CD8, IL-17A, IFN-γ, TNF-α, and NF-κB p65 mRNAs and decreased IL-4 mRNA in the arthritic control (AC) mice, while treatment of animals with AG126 reversed these effects. Western blot analysis confirmed the decreased expression of IL-17, GITR, NF-κB p65 proteins and increased Foxp3 and IL-4 proteins following AG126 treatment of knee tissue. Thus, our findings provide new evidence that inhibition of protein tyrosine kinase activity decreases the progression of arthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Blotting, Western , Cytokines/immunology , Female , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Tyrphostins/pharmacology
17.
J Biochem Mol Toxicol ; 30(11): 559-566, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27301782

ABSTRACT

In order to study the mechanisms underlying the alleviation of aflatoxin B1-induced genomic damage by proanthocyanidins (PAs), we examined the modulation of oxidative DNA damage induced by aflatoxin B1 in PAs-pretreated animals. The effects of PAs on changes in the expression of DNA damage and repair genes induced by aflatoxin B1 were also evaluated in rat marrow cells. Administration of PAs before aflatoxin B1 significantly mitigated aflatoxin B1-induced oxidative DNA damage in a dose-dependent manner. Aflatoxin B1 treatment induced significant alterations in the expression of specific DNA repair genes, and the pre-treatment of rats with PAs ameliorated the altered expression of these genes. Conclusively, PAs protect against aflatoxin B1-induced oxidative DNA damage in rats. These protective effects are attributed to the antioxidant effects of PA and enhanced DNA repair through modulation of DNA repair gene expression. Therefore, PAs are a promising chemoprotective agent for averting genotoxic risks associated with aflatoxin B1 exposure.


Subject(s)
Aflatoxin B1/toxicity , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , DNA Repair/drug effects , Proanthocyanidins/pharmacology , Aflatoxin B1/antagonists & inhibitors , Aflatoxin B1/isolation & purification , Animals , Aspergillus flavus/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Comet Assay , DNA Damage , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Male , Micronuclei, Chromosome-Defective , Micronucleus Tests , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Rats , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Life Sci ; 152: 1-13, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26997472

ABSTRACT

Aflatoxin B1 (AFB1) is immunotoxic to animals and is a suspected immunosuppressant in humans. ß-1,3-Glucan (BG) consists of glucose polymers and has a variety of stimulatory effects on the immune system. In this study, we investigated the role of BG on the expression of phenotypic markers and cytokine secretion in mice exposed to AFB1. We treated animals with BG (150mg/kg, p.o., once daily) for 7days beginning at the onset of AFB1 exposure. Exposure of animals to AFB1 alone (1250µg/kg, p.o, once daily) for 7days resulted in a decrease in the percentages of lymphocyte subsets (CD4(+), GITR(+), CD8(+), TCR ß(+), CD3(+), Foxp3(+), CD4(+)Foxp3(+), and CD127(+)) as compared to an normal control (NC). However, both BG alone and BG given in conjunction with exposure to AFB1 significantly increased the percentages of these lymphocyte subsets in blood. We also observed that mice exposed to AFB1 showed reduced IL-2, TNF-α, IL-17, and IFN-γ production in the spleen and serum. In contrast, oral administration of BG alone and in conjunction with AFB1 exposure augmented the levels of these cytokines. Moreover, this finding was confirmed through RT-PCR and western blot analysis of mRNA and protein expression in the spleen. Altogether, it can be concluded from these studies that BG enhances the responses of lymphocyte subsets, including cytokine production, even when given following exposure to AFB1 immunotoxin. These data demonstrate that BG carries out its immunomodulating activity by regulating cytokine production. Our findings also provide a direction for development of specific immunomodulating therapy.


Subject(s)
Aflatoxin B1/antagonists & inhibitors , Immunity/drug effects , Immunologic Factors/pharmacology , Immunosuppressive Agents/antagonists & inhibitors , beta-Glucans/pharmacology , Animals , Cytokines/blood , Cytokines/metabolism , Forkhead Transcription Factors/biosynthesis , Immunity, Cellular/drug effects , Lymphocyte Count , Lymphocyte Subsets/drug effects , Male , Mice , Mice, Inbred BALB C , Protein Synthesis Inhibitors/pharmacology , Spleen/drug effects , Spleen/metabolism , Transcription Factor RelA/biosynthesis
19.
J Neuroimmunol ; 289: 30-42, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26616869

ABSTRACT

Recently, the expression of histamine 4 receptor (H4R) on neurons was reported, however its function in cells within the central nervous system (CNS) remains poorly understood. To this end, we used the H4R agonist, 4-methylhistamine (4-MeH), and the H4R antagonist, JNJ77777120 (JNJ), to investigate the function of H4R signaling in immune cells in a murine model of chronic stress. Treatment of stressed mice with 4-MeH resulted in an increase in the proportion of lymphocyte subsets (CD3(+), CD8(+), CD28(+), and CD4(+)CD28(+)) and cells expressing the co-stimulatory molecules CD80(+) (B7.1) and CD86(+) (B7.2) in heparinized blood as compared to normal control (NC) and stressed control (SC) groups. We also observed that as compared to NC and SC mice, 4-MeH-treated mice showed greater production of IL-2(+), IL-6(+), IL-9(+), IL-21(+), and IL-27(+) cytokines in the spleen and by splenic CD4(+) T cells. Furthermore, 4-MeH treatment of stressed mice led to an increase in the levels of serum Th1/Th17 cytokines and corticosterone, and a decrease in Th2 cytokines. Treatment of chronically-stressed mice with 4-MeH also augmented expression of IL-6, IL-21, NF-κB p65, and STAT3 mRNA. Moreover, Western blot analyses confirmed increased protein expression of NF-κB, iNOS, and STAT3 expression following 4-MeH treatment of chronically-stressed mice as compared to controls. These proteins provide a novel relevant targets for the manipulation of chronic stress induced immune regulation. In striking contrast, treatment of stressed mice with the H4R antagonist, JNJ, resulted in a substantial reduction in all of the aforementioned effects upon immune cell percentages and cytokine production.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Cytokines/metabolism , Receptors, Histamine/metabolism , Signal Transduction/immunology , Stress, Psychological/immunology , Analysis of Variance , Animals , CD28 Antigens/genetics , Corticosterone/blood , Cytokines/genetics , Indoles/pharmacology , Male , Methylhistamines/pharmacology , Mice , Mice, Inbred BALB C , Piperazines/pharmacology , RNA, Messenger/metabolism , Restraint, Physical/psychology , Signal Transduction/drug effects , Spleen/metabolism , Spleen/pathology , Stress, Psychological/blood , Stress, Psychological/etiology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
20.
Asian Pac J Cancer Prev ; 16(14): 5807-15, 2015.
Article in English | MEDLINE | ID: mdl-26320455

ABSTRACT

BACKGROUND: Ovarian cancer is the most common gynecological malignancy and constitutes the fifth leading cause of female cancer death. Some biological parameters have prognostic roles in patients with advanced ovarian cancer and their expression may contribute to tumor progression. The aim of this study was to investigate the potential prognostic value of SKP2, genes P27Kip1, K-ras, c-Myc, COX2 and HER2 genes expression in ovarian cancer. MATERIALS AND METHODS: This study was performed on two hundred formalin fixed paraffin embedded ovarian cancer and normal adjacent tissues (NAT). Gene expression levels were assessed using real time PCR and Western blotting. RESULTS: Elevated expression levels of SKP2, K-ras, c-Myc, HER2 and COX2 genes were observed in 61.5% (123/200), 92.5% (185/200), 74% (148/200), 96 % (192/200), 90% (180/200) and 78.5% (157/200) of cancer tissues, respectively. High expression of SKP2 and down-regulation of P27 was associated with advanced stages of cancer. CONCLUSIONS: The association between high expression of c-Myc and SKP2 with low expression of P27 suggested that the Skp2-P27 pathway may play an important role in ovarian carcinogenesis. Reduced expression of P27 is associated with advanced stage of cancer and can be used as a biological marker in clinical routine assessment and management of women with advanced ovarian cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Ovarian Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , S-Phase Kinase-Associated Proteins/genetics , Tumor Cells, Cultured , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...