Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Toxicology ; 504: 153788, 2024 May.
Article in English | MEDLINE | ID: mdl-38527609

ABSTRACT

Cisplatin is a highly effective chemotherapy drug used to treat most solid tumors. However, one of its side effects is testicular toxicity, which can lead to fertility abnormalities. This study investigated the effectiveness of dental pulp mesenchymal stem cells conditioned medium (DPSC-CM) on cisplatin-induced testicular toxicity. In this study, 36 eight-week-old male Wistar rats were randomly divided into three groups equally (n = 12). Group 1 control "CTR", which received normal saline (0.5 ml) intraperitoneally (i.p), group 2 "Cis" which received an intraperitoneal dose of cisplatin (7 mg/kg), and group 3 "Cis+CM" which received an i.p injection of DPSC-CM (0.5 mg/kg) after cisplatin injection. Biochemical, histomorphometric, and histopathological studies were performed on the testis. Our results exhibited that cis administration led to a decline in total body weight, testis weight, diameter, and volume. A decrease in testosterone and IL-6 serum levels, as well as a decrease in IL-6 and TNFα levels, the activity of catalase and SOD enzymes, and an increase in MDA in testicular tissue were detected. Testicular tissue damage was associated with a significant decrease in tube diameter, germinal epithelium height, number of spermatogonia and Sertoli cells, along with a noticeable increase in basement membrane thickness, and perivascular fibrosis. DMSC-CM improved all the mentioned parameters. Taken together, our results demonstrated that DMSC-CM due to its antioxidant and anti-inflammatory properties, could be effective in reversing cisplatin-induced testicular toxicity.


Subject(s)
Cisplatin , Dental Pulp , Rats, Wistar , Testis , Animals , Male , Cisplatin/toxicity , Culture Media, Conditioned/pharmacology , Testis/drug effects , Testis/pathology , Testis/metabolism , Dental Pulp/drug effects , Dental Pulp/cytology , Rats , Testosterone/blood , Antineoplastic Agents/toxicity , Oxidative Stress/drug effects , Mesenchymal Stem Cells/drug effects
2.
J Oral Biol Craniofac Res ; 13(2): 155-163, 2023.
Article in English | MEDLINE | ID: mdl-36618007

ABSTRACT

Background: Amygdalin is known as a chemical compound derived from various fruits. The glycosides existing in this plant have been historically utilized as an anticancer agent. This review presented an overview of amygdalin and its onco-immunity and other therapeutic medical applications. Method: A literature search for studies relating to amygdalin and cancer treatment was carried out using PubMed and Google Scholar. Combinations of the following terms were used in the search strategies: "amygdalin," "rhodanese," "cyanide," "cyanogenic," "hypothiocyanite," "mandelonitrile," "glucosides," "cancer," "apoptosis," and "cytotoxicity," combined with a cancer term such as "seed," "almond," or "apricot," "cancer + cell line, antiproliferation or inhibition," "BAX From the March 3, 1981 until the April 15, 2021, all of the English-language papers were evaluated based on the inclusion criteria. Publications included reviews, chapters from books, and original research papers. Results: The FDA prohibits Amygdalin from medical usage as an anticancer treatment due to a lack of proof of cure in cancer cases. When this natural-based compound is used with conditional chemotherapeutic medicines causes synergistic effects. Besides, amygdalin is used to manage asthma, improve the immune system, induce apoptosis in human renal fibroblasts, and inhibit hyperglycemia. Conclusion: Various medical uses of amygdalin have been found such as managing asthma, improving the immune system, inducing apoptosis in human renal fibroblasts, and inhibiting hyperglycemia. More effective in vitro and review studies are required to elucidate the exact role of this herb in medical applications.

3.
Stem Cell Rev Rep ; 19(2): 322-344, 2023 02.
Article in English | MEDLINE | ID: mdl-36272020

ABSTRACT

The breakdown of self-tolerance of the immune response can lead to autoimmune conditions in which chronic inflammation induces tissue damage. Systemic lupus erythematosus (SLE) is a debilitating multisystemic autoimmune disorder with a high prevalence in women of childbearing age; however, SLE incidence, prevalence, and severity are strongly influenced by ethnicity. Although the mystery of autoimmune diseases remains unsolved, disturbance in the proportion and function of B cell subsets has a major role in SLE's pathogenesis. Additionally, colocalizing hyperactive T helper cell subgroups within inflammatory niches are indispensable. Despite significant advances in standard treatments, nonspecific immunosuppression, the risk of serious infections, and resistance to conventional therapies in some cases have raised the urgent need for new treatment strategies. Without the need to suppress the immune system, mesenchymal stem cells (MSCs), as ''smart" immune modulators, are able to control cellular and humoral auto-aggression responses by participating in precursor cell development. In lupus, due to autologous MSCs disorder, the ability of allogenic engrafted MSCs in tissue regeneration and resetting immune homeostasis with the provision of a new immunocyte repertoire has been considered simultaneously. In Brief The bone marrow mesenchymal stem cells (BM-MSCs) lineage plays a critical role in maintaining the hematopoietic stem-cell microstructure and modulating immunocytes. The impairment of BM-MSCs and their niche partially contribute to the pathogenesis of SLE-like diseases. Allogenic MSC transplantation can reconstruct BM microstructure, possibly contributing to the recovery of immunocyte phenotype restoration of immune homeostasis. In terms of future prospects of MSCs, artificially gained by ex vivo isolation and culture adaptation, the wide variety of potential mediators and mechanisms might be linked to the promotion of the immunomodulatory function of MSCs.


Subject(s)
Lupus Erythematosus, Systemic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/therapy , Cell Differentiation , Immunosuppression Therapy , Mesenchymal Stem Cells/metabolism
4.
Stem Cell Rev Rep ; 18(8): 2781-2796, 2022 12.
Article in English | MEDLINE | ID: mdl-35876958

ABSTRACT

Lupus is known as a systemic immune-mediated disorder. Like other diseases in this category, its cause and definitive treatment remain unknown. Gold standard therapies, which mainly include immunosuppressive agents, have been able to have therapeutic effects on patients. However, a significant percentage of cases still do not respond to this kind of treatment, resulting in death from complications. Recently, a new source of non-hematopoietic cells, mesenchymal stem/stromal cells (MSCs), with the potency to re-establishment immune homeostasis and tissue regeneration, has been wildly used in both primary and clinical research. One of the remarkable features of MSCs is their anti-inflammatory and immunosuppressive properties and stimulating tissue differentiation programs. Under the influence of background signals, MSCs migrate to inflammatory bioactive substances and then regulate overactive immune responses to restore immune tolerance. MSCs have shown a two-way interaction with most immunocytes, which plays a significant role in resolving sterile inflammation. Restricting the entry of inflamed cells into the site of inflammation and re-educated infiltrated cells to achieve a tolerant phenotype have been reported as mechanisms of MSCs in tissue repair. Stimulation of the endogenous and tissue-dwelling stem cells in addition to releasing immunomodulatory agents, suggests MSCs transplantation as a potential modality in the treatment of future immune-mediated disorders.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cell Transplantation/methods , Immune Tolerance , Wound Healing , Inflammation
5.
Biomed Res Int ; 2022: 4767621, 2022.
Article in English | MEDLINE | ID: mdl-35637752

ABSTRACT

Cancer of the prostate is an indicated type that is often recorded as a kind of cancer in men and the second critical cause of mortality through cancer cases. Many pharmacological investigations have shown that numerous herbal substances possess anticancer action. Amygdalin (AMD) has antitumour capabilities and works as an antioxidant, antibacterial, anti-inflammatory, and immune-regulating characteristics. The anticancer effects of amygdalin and its metabolizing enzymes, rhodanese (RHD) and betaglucosidase (BGD), were examined in vivo, as well as their antitumour processes. Novel, effective combination agents are necessary to increase existing cancer treatment rates. This research was aimed at determining the anticarcinogenic impact of amygdalin (AMD) in vivo. This research was aimed at determining the RHD and BGD on the anticarcinogenic impact of AMD in vivo. Subcutaneously, PC3 prostate cancer cell lines were implanted into nude mice. Mice were treated every day with 0.5 ml of 50 mg/ml (AMD), AMD+ (RHD 0.1 mg/ml), AMD+(BGD 0.1 mg/ml), and doxorubicin (DOX 50 mg/ml). Mice were normalized for negative control with untreated mice. In in vivo, morphopathological alterations in the tumour tissue were analyzed by histopathological staining methods. After 35 days of therapy, tumour growth and size inhibition were evident, indicating a function for the metabolic enzymes BGD and RHD in regulating AMD's anticancer effect in vivo. We concluded the critical role of metabolic enzymes BGD and RHD in elevating the antigrowth of PC3 cancer cell lines in Balb/c nude mice treated with AMD.


Subject(s)
Adenocarcinoma , Amygdalin , Prostatic Neoplasms , Adenocarcinoma/drug therapy , Amygdalin/pharmacology , Amygdalin/therapeutic use , Animals , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Humans , Male , Mice , Mice, Nude , Prostate/pathology , Prostatic Neoplasms/pathology
6.
Article in English | MEDLINE | ID: mdl-34503439

ABSTRACT

BACKGROUND: Many investigations have expanded this concept that liver chronic inflammation has an essential role in persistent cell damages along with altering the liver microenvironment leading to fibrosis, cirrhosis, and finally, hepatocellular carcinoma (HCC). To reduce inflammation and relieve symptoms, Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) are commonly used; however, their long-term usage can lead to severe adverse events on vital organs like the liver. Interestingly, the α-L-Guluronic Acid (G2013), as a novel NSAID with immunomodulatory properties, has shown the inhibitory effects on inflammation and metastasis in experimental models. OBJECTIVE: This study was conducted to determine the effects of G2013 on cytotoxicity and induction of apoptosis, as a new therapeutic target for cancer therapy, in the HepG2 cell line and the mouse fibroblast cell line L929, as a control. METHODS: MTT assay and flow cytometry method were carried out using the different concentrations of G2013 (5, 15, 25, 50, 100, 200 and 400 µg/ml) in 3 distinct incubation times. RESULTS: Our data showed that treatment of HepG2 cells with high concentration (400µg/mL) of G2013 could effectively cause a decrease in cell viability, so that they were statistically different after 72 hours compared to other concentrations (5 to 200 µg/ml) (p<0.05 and p<0.01, respectively). Moreover, the proportion of apoptosis of HepG2 cells at the dose of 200µg/mL considerably increased, suggesting that the induction of apoptosis by G2013 in HepG2 cells is dose- and time-dependent, which could promote its anticancer properties. CONCLUSION: The present study revealed that G2013 could induce apoptosis in the liver cancer model. Therefore, based on these findings, G2013 might be considered as a therapeutic option in cancer therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Line , Hexuronic Acids , Liver Neoplasms/drug therapy , Mice , Tumor Microenvironment
7.
Curr Rheumatol Rev ; 17(4): 404-411, 2021.
Article in English | MEDLINE | ID: mdl-33687897

ABSTRACT

OBJECTIVES: The present study was designed to evaluate the association of transporters associated with antigen processing (TAP2) polymorphisms TAP2-379Ile > Val (rs1800454), TAP2-665Thr > Ala (rs241447) and TAP2-565Ala > Thr (rs2228396) as a candidate gene with susceptibility to the Systemic Lupus Erythematosus (SLE). METHODS: To analyze these three polymorphic variants, 88 patients with SLE and 100 healthy controls from northeastern Iran were enrolled from May 2018 to July 2019. Genomic DNA polymorphisms were performed by amplification refractory mutation system-polymerase chain reaction (ARMS-PCR) technique. Data were analyzed by SPSS software. RESULTS: In this cross-sectional study, there was a stratification between patients and controls. The distribution of the frequency of Ala73 (41.5%) allele at TAP2/665 and Ile 19 (10.8%) allele at TAP2/379 was higher in patients. Additionally, the Ala/Ala 13(14.8%) and Ala/Thr 49(55.7%) genotypes distributions at 665 positions were higher in SLE patients compared to the controls. Furthermore, frequencies of TAP2*H allele significantly increased in SLE patients 10(5.71%) (P=0.01). Frequency of TAP2*A allele in the control group was 120(60%) (p=0.06) due to the dominant genetic model. This allele has a protective effect against SLE. There was no relationship between TAP2*D, TAP2*E, TAP2*F and TAP2*G alleles with the outbreak of SLE. CONCLUSION: Our data indicated that genetic variants in TAP2 gene may be associated with SLE disease. A correlation between Ala allele at TAP2/665 and Ile allele at TAP2/379 polymorphisms and pathogenesis of SLE was observed.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 3 , Lupus Erythematosus, Systemic , Polymorphism, Genetic , ATP Binding Cassette Transporter, Subfamily B, Member 3/genetics , Case-Control Studies , Cross-Sectional Studies , Genetic Predisposition to Disease , Humans , Lupus Erythematosus, Systemic/genetics , Polymorphism, Genetic/genetics
8.
Adv Exp Med Biol ; 1286: 87-105, 2021.
Article in English | MEDLINE | ID: mdl-33725347

ABSTRACT

Gut microbiota has essential roles in the prevention and progression of multiple sclerosis (MS). The association between the gut microbiota and the central nervous system (CNS) or immune system response of MS patients has been documented in many studies. The composition of the gut microbiota could lead to sensitization or resistance against promotion and development of MS disease. Probiotics are the major part of gut microflorapopulation and could be substituted with tolerogenic probiotics that protect the CNS against autoimmune responses. Tolerogenic probiotics with anti-inflammatory and immuno-modulatory properties have effects on intestinal flora and can reestablish regulatory mucosal and systemic immune responses. Probiotics are able to prevent and restore excessive activation of inflammatory responses, especially autoreactive T cells and inflammatory cytokines. Tolerogenic probiotics, through induction of regulatory T cells and increase of anti-inflammatory cytokines, play a crucial role in controlling inflammation and maintaining tolerance and hemostasis. Therefore, probiotics can be considered as a preventive or therapeutic tool in MS. In the present review, we focus on the immunoregulatory effects of tolerogenic probiotics on the severity of disease, as well as Th1, Th2, and Treg populations in different experimental and human studies of MS.


Subject(s)
Gastrointestinal Microbiome , Multiple Sclerosis , Probiotics , Cytokines , Humans , Immune Tolerance , Multiple Sclerosis/drug therapy , Probiotics/therapeutic use
9.
J Cell Mol Med ; 24(23): 13573-13588, 2020 12.
Article in English | MEDLINE | ID: mdl-33135395

ABSTRACT

Autoreactive inflammatory CD4+ T cells, such as T helper (Th)1 and Th17 subtypes, have been found to associate with the pathogenesis of autoimmune disorders. On the other hand, CD4+ Foxp3+ T regulatory (Treg) cells are crucial for the immune tolerance and have a critical role in the suppression of the excessive immune and inflammatory response promoted by these Th cells. In contrast, dendritic cells (DCs) and macrophages are immune cells that through their inflammatory functions promote autoreactive T-cell responses in autoimmune conditions. In recent years, there has been increasing attention to exploring effective immunomodulatory or anti-inflammatory agents from the herbal collection of traditional medicine. Berberine, an isoquinoline alkaloid, is one of the main active ingredients extracted from medicinal herbs and has been shown to exert various biological and pharmacological effects that are suggested to be mainly attributed to its anti-inflammatory and immunomodulatory properties. Several lines of experimental study have recently investigated the therapeutic potential of berberine for treating autoimmune conditions in animal models of human autoimmune diseases. Here, we aimed to seek mechanisms underlying immunomodulatory and anti-inflammatory effects of berberine on autoreactive inflammatory responses in autoimmune conditions. Reported data reveal that berberine can directly suppress functions and differentiation of pro-inflammatory Th1 and Th17 cells, and indirectly decrease Th cell-mediated inflammation through modulating or suppressing other cells assisting autoreactive inflammation, such as Tregs, DCs and macrophages.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autoimmune Diseases/etiology , Autoimmunity/drug effects , Berberine/pharmacology , Immunologic Factors/pharmacology , Inflammation/etiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/etiology , Autoimmune Diseases of the Nervous System/metabolism , Cytokines/biosynthesis , Demyelinating Autoimmune Diseases, CNS/diagnosis , Demyelinating Autoimmune Diseases, CNS/etiology , Demyelinating Autoimmune Diseases, CNS/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunomodulation/drug effects , Inflammation/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism
10.
Article in English | MEDLINE | ID: mdl-32807059

ABSTRACT

The article has been withdrawn by agreement between the editors and publisher of Current Drug Discovery Technologies. The authors are not responding to the editor's requests to provide the language-edited version.Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php Bentham Science Disclaimer: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript, the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

11.
Atherosclerosis ; 285: 1-9, 2019 06.
Article in English | MEDLINE | ID: mdl-30939341

ABSTRACT

The stem cell-based therapy has emerged as a promising therapeutic strategy for treating cardiovascular ischemic diseases (CVIDs), such as myocardial infarction (MI). However, some important functional shortcomings of stem cell transplantation, such as immune rejection, tumorigenicity and infusional toxicity, have overshadowed stem cell therapy in the setting of cardiovascular diseases (CVDs). Accumulating evidence suggests that the therapeutic effects of transplanted stem cells are predominately mediated by secreting paracrine factors, importantly, microRNAs (miRs) present in the secreted exosomes. Therefore, novel cell-free therapy based on the stem cell-secreted exosomal miRs can be considered as a safe and effective alternative tool to stem cell therapy for the treatment of CVDs. Stem cell-derived miRs have recently been found to transfer, via exosomes, from a transplanted stem cell into a recipient cardiac cell, where they regulate various cellular process, such as proliferation, apoptosis, stress responses, as well as differentiation and angiogenesis. The present review aimed to summarize cardioprotective exosomal miRs secreted by transplanted stem cells from various sources, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and cardiac stem/progenitor cells, which showed beneficial modulatory effects on the myocardial infracted heart. In summary, stem cell-exosomal miRs, including miR-19a, mirR-21, miR-21-5p, miR-21-a5p, miR-22 miR-24, miR-26a, miR-29, miR-125b-5p, miR-126, miR-201, miR-210, and miR-294, have been shown to have cardioprotective effects by enhancing cardiomyocyte survival and function and attenuating cardiac fibrosis. Additionally, MCS-exosomal miRs, including miR-126, miR-210, miR-21, miR-23a-3p and miR-130a-3p, are found to exert cardioprotective effects through induction of angiogenesis in ischemic heart after MI.


Subject(s)
Cardiovascular Diseases/drug therapy , Exosomes/genetics , MicroRNAs/therapeutic use , Animals , Cardiotonic Agents/therapeutic use , Exosomes/metabolism , Humans , MicroRNAs/metabolism , Stem Cells/metabolism , Stem Cells/ultrastructure
12.
J Clin Pediatr Dent ; 42(3): 203-207, 2018.
Article in English | MEDLINE | ID: mdl-29698133

ABSTRACT

OBJECTIVES: The purpose of the present study was to evaluate the efficacy of Ham's F-10 in maintaining the viability and reproducibility of PDL cells on avulsed teeth. STUDY DESIGN: Sixty mature, healthy extracted premolars were used. The experimental media used were Ham's F-10, Hank's balanced salt solution (HBSS), skim milk, and tap water (n = 15 specimens each). Cell viability was tested after 1, 3, 6, and 24 h storage in medium. Cell reproducibility was assessed by methyl-thiazol-tetrazolium (MTT) assay after1, 3, and 6 h storage in Ham's F-10, HBSS, and tap water. RESULTS: The viability of PDL cells stored in Ham's F-10 and HBSS was significantly greater than that of samples stored in milk and tap water at all-time points (P<0.001). A significant difference in cell viability between samples stored in Ham's F-10 and HBSS (favoring the former) was observed only at 6h (P=0.04). MTT assay results were significantly better for samples stored in Ham's F-10 and HBSS than for those stored in tap water (P<0.001), with a significant difference between Ham's F-10 and HBSS observed only at 3h (P<0.001). CONCLUSIONS: Ham's F-10 is capable of preserving PDL cells viable and reproducible better than milk and tap water and similar to HBSS.


Subject(s)
Organ Preservation Solutions , Periodontal Ligament/cytology , Cell Proliferation , Cell Survival , Humans , Tooth Avulsion
13.
Eur J Pharm Sci ; 47(2): 474-80, 2012 Sep 29.
Article in English | MEDLINE | ID: mdl-22750628

ABSTRACT

In this study we aimed to evaluate PXR and ABCG2 gene expression patterns and NF-κB activity induced by proinflammatory cytokines in different breast normal and carcinoma cells. The effects of proinflammatory cytokines on ABCG2 and PXR mRNA expression were studied using real-time PCR. Western blot analysis used for evaluating the protein levels of ABCG2, PXR and the active form of NF-κB (p65 in nuclear protein extract). Significant inductions in the ABCG2 and PXR mRNA and protein levels and NF-κB activity, were observed in MCF7, BT-474, CAL51, 184A1 and HBL100 cells, upon treatment with 50 ng/ml of IL-1ß and TNF-α. On the contrary significant reduction of the ABCG2 and PXR mRNA and protein levels and NF-κB activity, were observed in MDA-MB-435 cell line. In conclusion, IL-1ß and TNF-α induced ABCG2 and PXR expression and NF-κB activity in some breast cancer and normal cell lines. Similar patterns of induction and reduction in PXR and ABCG2 genes and NF-κB activity suggest a probable relationship between ABCG2, PXR and NF-κB.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Receptors, Steroid/metabolism , Tumor Necrosis Factor-alpha/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Female , Humans , Neoplasm Proteins/genetics , Pregnane X Receptor , RNA, Messenger/metabolism , Receptors, Steroid/genetics
14.
Mol Cell Biochem ; 363(1-2): 385-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22193459

ABSTRACT

The ATP-binding cassette sub-family G member 2 (ABCG2) is implicated as a member of multidrug resistant proteins in tumors, mediating efflux of a wide spectrum of anticancer drugs. Pro-inflammatory cytokines, which are present within the micro-environment of tumors and inflammation, are able to modulate the expressions and activities of different drug transporters. This study was aimed to evaluate the short-term (72-h treatment) effects of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) on the expression and function of ABCG2 in cervix carcinoma and gastric cancer cells. Effects of pro-inflammatory cytokines on mRNA, protein expression, and function of ABCG2 were studied using real time RT-PCR and flow cytometry methods, respectively. HeLa cells treated with IL-1ß, IL-6, or TNF-α showed decrements in ABCG2 mRNA levels without any changes in protein expression and function of ABCG2. IL-6 and TNF-α had no effects on mRNA, protein expression, and function of ABCG2 in EPG85-257 cells. Although IL-1ß did not alter ABCG2 at mRNA or protein levels in EPG85-257 cells, it augmented function of ABCG2 in these cells. Mitoxantrone accumulation was also amplified in IL-1ß-, IL-6- or TNF-α-treated HeLa cells and in IL-1ß-treated EPG85-257 cells. In conclusion, pro-inflammatory cytokines were able to modulate the expression of ABCG2 at transcriptional and post-transcriptional levels in human cervix and gastric cancer cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uterine Cervical Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents/metabolism , Drug Resistance, Neoplasm , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Mitoxantrone/metabolism , Neoplasm Proteins/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Time Factors , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology
15.
DNA Cell Biol ; 30(1): 25-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20846001

ABSTRACT

In this study, we aimed to evaluate the relationship between PXR and ABCG2 gene expression patterns induced by proinflammatory cytokines in MCF7 and MCF7/MX breast carcinoma cell lines. The effects of proinflammatory cytokines on ABCG2 and PXR mRNA expression were studied using the real-time polymerase chain reaction method. Significant inductions in the ABCG2 and PXR mRNA levels were observed in MCF7 cells, upon treatment with interleukin-1ß and tumor necrosis factor-α, whereas MCF7/MX cells did not significantly respond to the treatment. The results also show that in comparison to MCF7 cell line the basal mRNA expression level of PXR was higher in the MCF7/MX cell line. In conclusion, interleukin-1ß and tumor necrosis factor-α induced ABCG2 and PXR mRNAs in the MCF7 breast cancer cell line; no significant changes on expression of the same genes in MCF7/MX cells were observed. This differential effect of the cytokines on two different cell lines seems to be influenced by basal levels of the mRNAs, which are very high in MCF7/MX cells. Similar patterns of induction in PXR and ABCG2 genes suggest a probable relationship between these two factors.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Breast Neoplasms/pathology , Cytokines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/genetics , Receptors, Steroid/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Inflammation/metabolism , Pregnane X Receptor , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Rheumatol Int ; 30(11): 1537-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20473502

ABSTRACT

Several data link vitamin D to many autoimmune diseases. Association between vitamin D receptor (VDR) gene BsmI polymorphisms and systemic lupus erythematosus has been reported. In this study, we examine whether the VDR gene BsmI polymorphisms are markers for susceptibility to or severity of SLE. This study incorporated 60 patients with SLE living in northeastern Iran. Three genotypes, BB, Bb and bb, were detected based on polymerase chain reaction-restriction fragment length polymorphism (PCR/RFLP). The distribution of VDR genotyping in patients with SLE was 23.3% for BB, 60% for Bb and 16.7% for bb and in the control group was 33.3% for BB, 46.7% for Bb and 20% for bb (P = 0.334). No association of VDR gene BsmI polymorphisms with SLE disease activity index (SLEDAI), SLE damage score and major organ involvement was detected. The result of this study did not show any association between VDR gene BsmI polymorphisms and SLE.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Receptors, Calcitriol/genetics , Adult , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Humans , Iran/epidemiology , Lupus Erythematosus, Systemic/epidemiology , Lupus Erythematosus, Systemic/metabolism , Male , Young Adult
17.
Iran J Pediatr ; 20(1): 41-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-23056680

ABSTRACT

OBJECTIVE: Late-onset sepsis is responsible for high morbidity and mortality in newborn infants in the world and in particular in developing countries. In this study, we evaluated whether clinical characteristics, laboratory parameters and measurements of serum interleukin-8 (IL-8) are able to discriminate between late neonatal sepsis and normal baby. METHODS: This was a prospective (case-control) study conducted between March 2007 and April 2008, at the neonatal intensive care unit, Ghaem Hospital, Mashhad, Iran. The study comprised 93 neonates ≥72 hours of life. The infants were categorized in two groups based on the clinical presentation, and biochemical markers including complete blood count, C-reactive protein (CRP) and blood culture: 1) Control group including 42 infants with routine screening and 2) Case group consisting of 38 infants with definitive infection (positive blood and/or cerebrospinal fluid culture) or clinical sepsis (clinical and laboratory signs of infection without positive blood or CSF culture). Receiver-operating characteristic curves were used for the determination of thresholds for the infection group versus healthy neonate group. FINDINGS: Eighty infants were enrolled in this study. IL-8 and CRP decreased in order of definitive infection, clinical sepsis and healthy subjects respectively (P<0.001). Sensitivity, specificity, positive predictive value, negative predictive value for serum levels were 0.95, 0.1, 0.97, 0.1 for IL-8 and 0.83, 0.86, 0.83, 0.69 for CRP respectively (cut-off point for IL-8 >60pg/ml and for CRP>6mg/dl). CONCLUSION: IL-8 may be a valid and early predictive marker of neonatal infection. Also, IL-8 is associated with severity of infection.

18.
Inflamm Res ; 58(10): 669-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19333723

ABSTRACT

OBJECTIVE: In this study, we aimed to evaluate the influence of proinflammatory cytokines on ABCG2 expression and function in human MCF-7 breast cancer cell line and its mitoxantrone-resistant derivative MCF-7/MX. METHODS: The effects of proinflammatory cytokines on ABCG2 mRNA expression were studied using real-time PCR method. Cytokine-mediated modification of ABCG2 protein expression and function was investigated by means of flow cytometry. RESULTS: Significant inductions in the ABCG2 mRNA levels, protein expression, and activity were observed in IL-1 beta and TNF-alpha-treated MCF-7 cells. IL-6 increased ABCG2 protein, but had no effects on ABCG2 mRNA and function in MCF-7 cells. Although IL-1 beta did not alter mRNA and protein levels of the transporter in MCF-7/MX cells, ABCG2-mediated efflux was significantly increased in IL-1 beta-treated MCF-7/MX cells. TNF-alpha-treated MCF-7/MX cells also demonstrated greater ABCG2 protein expression and function without any changes in mRNA levels of the transporter. Neither ABCG2 mRNA nor its protein expression and function were affected by IL-6 in MCF-7/MX cells. CONCLUSION: IL-1 beta and TNF-alpha induce ABCG2 mRNA and protein expression and increase its activity in breast cancer cell line MCF-7. In MCF-7/MX cells these cytokines modulate ABCG2 protein expression and/or function, but they have no influence on the transporter mRNA levels.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Interleukin-1beta/physiology , Mitoxantrone , Neoplasm Proteins/metabolism , Tumor Necrosis Factor-alpha/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/pharmacology , Mitoxantrone/therapeutic use , Neoplasm Proteins/genetics , RNA, Messenger/metabolism
19.
Rheumatol Int ; 28(2): 189-91, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17763852

ABSTRACT

There are controversial reports that TNF-a promoter polymorphism may be an independent marker of susceptibility to rheumatoid arthritis (RA). We used Polymerase chain reaction amplification and Restriction fragment length polymorphism for analysis of the polymorphism at position -308 in promoter of TNF-alpha gene in 34 patients with RA and 30 healthy individuals. Distribution of TNF-genotypes in RA patients did not differ from that in controls. Moreover, there was apparent association between the -308 TNF-alpha polymorphism and erosions in hand x-Ray was found (P value = 0.043). We suggest that TNF-alpha -308 promoter polymorphism is not a genetic risk factor for RA susceptibility but may be associated with radiographic damage in rheumatoid patients.


Subject(s)
Arthritis, Rheumatoid/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/genetics , Alleles , Case-Control Studies , DNA/genetics , DNA/isolation & purification , DNA Primers/chemistry , Gene Frequency , Genetic Predisposition to Disease , Hand/diagnostic imaging , Hand/pathology , Humans , Nucleic Acid Amplification Techniques , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Radiography , Risk Factors , Severity of Illness Index , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...