Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 114: 109568, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36527883

ABSTRACT

Cancer is a disease caused when cells divide uncontrollably and spread into surrounding tissues. There are different therapeutic modalities that control cancer growth, of which surgery, chemotherapy, and radiotherapy. Chemotherapy is a cancer treatment approach in which medications are used to inhibit cell proliferation and tumor multiplication, thus avoiding invasion and metastasis and thus eradicate cancer. One of the common complications associated with cancer chemotherapy is rapid lysis of expanding tumor cells, known as tumor lysis syndrome (TLS). TLS is associated with number of metabolic changes such as hyperuricemia, hyperkalemia, hyperphosphatemia and hypocalcemia. Among the consequences of hyperuricemia, hyperkalemia, hyperphosphatemia and hypocalcemia is the inhibition of 5' AMP-activated protein kinase (AMPK). Inhibition of AMPK induced different cancer chemo-resistance mechanisms such as cancer stem cells (CSCs), p-glycoproteins, Octamer-binding transcription factor 4 (OCT-4), homeobox protein NANOG, Krüppel-like factor 4 (KLF4) and immune microenvironment and thus leads to poor response to chemotherapy and even relapses after treatment. Our review aims to uncover new mechanisms underlying the metabolic consequences of tumor lysis on AMPK in tumor microenvironment. In this review, we also investigated the effect of AMPK on different cancer chemo-resistance mechanisms such as cancer stem cells, p-glycoproteins, OCT-4, NANOG, KLF4 and immune microenvironment.


Subject(s)
Hyperkalemia , Hyperphosphatemia , Hyperuricemia , Hypocalcemia , Tumor Lysis Syndrome , Humans , AMP-Activated Protein Kinases , Drug Resistance, Neoplasm , Hyperkalemia/complications , Hyperkalemia/therapy , Hyperphosphatemia/complications , Hypocalcemia/complications , Hypocalcemia/therapy , Neoplasm Recurrence, Local , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/therapy , Tumor Microenvironment , Protein Kinase Inhibitors/therapeutic use
2.
Int Immunopharmacol ; 113(Pt B): 109475, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36435064

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common type of liver cancer. There are different therapeutic approaches to cancer eradication such as chemotherapy, radiotherapy, and surgery. The fuel for treatment resistance is heterogeneity, which also has significant effects on cancer therapies and biomarker research. One of the most common causes of cancer chemoresistance and relapse is the presence and percentage of cancer stem cells (CSCs), among them CD47+ CSCs. Besides, the change in the tumor microenvironment (TME) stands as one of the main factors for the failure of chemotherapeutic protocols. The current review aims to focus on how the change in immune mediators such as TGF-ß, IL-10, IL-17, IDO, Gal-1, PD-L1, and CTLA-4 affect CD47+ CSCs in HCC and thus open a new era towards developing new approaches for prevention of HCC relapse and stemness through different immune modulation approaches. Then we investigate some drugs that have a dual effect on both TME and CD47 CSCs and thus the best choice in comorbidities.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/therapy , CD47 Antigen , Liver Neoplasms/drug therapy , Neoplasm Recurrence, Local , Neoplastic Stem Cells , Tumor Microenvironment
3.
J Transl Med ; 20(1): 544, 2022 11 26.
Article in English | MEDLINE | ID: mdl-36435779

ABSTRACT

Liver fibrosis is a liver disease in which there is an excessive buildup of extracellular matrix proteins, including collagen. By regulating cytokine production and the inflammatory response, heat shock proteins (HSPs) contribute significantly to a wider spectrum of fibrotic illnesses, such as lung, liver, and idiopathic pulmonary fibrosis by aiding in the folding and assembly of freshly synthesized proteins, HSPs serve as chaperones. HSP70 is one of the key HSPs in avoiding protein aggregation which induces its action by sending unfolded and/or misfolded proteins to the ubiquitin-proteasome degradation pathway and antagonizing influence on epithelial-mesenchymal transition. HSP47, on the other hand, is crucial for boosting collagen synthesis, and deposition, and fostering the emergence of fibrotic disorders. The current review aims to provide light on how HSP70 and HSP47 affect hepatic fibrogenesis. Additionally, our review looks into new therapeutic approaches that target HSP70 and HSP47 and could potentially be used as drug candidates to treat liver fibrosis, especially in cases of comorbidities.


Subject(s)
HSP47 Heat-Shock Proteins , Heat-Shock Proteins , Humans , HSP47 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Fibrosis , Liver Cirrhosis/drug therapy , Collagen/metabolism
4.
J Transl Med ; 20(1): 347, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35918736

ABSTRACT

Cancer is one of the leading causes of death in both men and women worldwide. One of the main changes associated with cancer progression, metastasis, recurrence, and chemoresistance is the change in the tumor immune microenvironment, especially immunosuppression. Cancer immunosuppression appears in multiple forms, such as inhibition of immuno-stimulant cells with downregulation of immuno-stimulant mediators or through stimulation of immuno-suppressive cells with upregulation of immunosuppressive mediators. One of the most immunosuppressive mediators that approved potency in lung cancer progression is indoleamine 2,3-dioxygenase (IDO) and its metabolite kynurenine (Kyn). The current review tries to elucidate the role of IDO/Kyn on cancer proliferation, apoptosis, angiogenesis, oxidative stress, and cancer stemness. Besides, our review investigates the new therapeutic modalities that target IDO/Kyn pathway and thus as drug candidates for targeting lung cancer and drugs that potentiate IDO/Kyn pathway and thus can be cancer-promoting agents.


Subject(s)
Kynurenine , Lung Neoplasms , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Male , Tryptophan/metabolism , Tumor Microenvironment
5.
Int Immunopharmacol ; 109: 108889, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35679661

ABSTRACT

Hyperinsulinemia, hyperglycemia, and chronic inflammation may play a role in hepatocellular carcinoma (HCC). Treatment of HCC patients with the antidiabetic medication metformin corrected the pathological changes of HCC by affecting proliferation, apoptosis, and angiogenesis. On the other hand, our review aims to uncover new pathways underlying metformin's anti-tumor action in the liver, focusing on immunological mediators and immune archetypes. In this review, we discuss the effect of metformin on restructuring the HCC immune microenvironment, such as dendritic cells, T cells, Tregs, macrophages, neutrophils, and myeloid-derived suppressors cells (MDSCs). Furthermore, Metformin also changes the expression pattern of immune mediators in HCC immune microenvironment, including programmed cell death 1 (PD-1), cytotoxic T lymphocyte antigen-4 (CTLA-4), transforming growth factor-beta (TGF-ß), Interleukin 12 (IL-12), indoleamine 2,3 dioxygenase (IDO), forkhead box protein P3 (FOXP3), and interferon-gamma (IFN-γ). This review summarizes a state-of-the-art understanding of the molecular mechanisms underlining novel anticarcinogenic approaches of metformin through modulation of liver cancer immune microenvironment both on the cellular and molecular scales, which aids in regaining immune fitness and thus better prognosis. The changes in tumor immune architecture and mediators induced by metformin make it a robust antineoplastic agent with multiple mechanisms of action, especially for people with diabetes and HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , Humans , Metformin/pharmacology , Metformin/therapeutic use , Tumor Microenvironment
6.
Biomed Pharmacother ; 147: 112628, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35032769

ABSTRACT

Acute lung injury (ALI) is one the most common causes of morbidity and mortality in critically ill patients. In this study, we examined for first time the role of dapagliflozin (DPGZ) in lipopolysaccharide (LPS)-induced ALI in rats and determined the underlying molecular mechanisms by evaluating the effects of DPGZ on adenosine monophosphate kinase (AMPK), nuclear transcription factor kappa B, nucleotide-binding and oligomerization domain-like receptor 3 inflammasome activation. Treatment of acute lung injured rats with either low dose (5 mg/kg) or high dose (10 mg/kg) DPGZ significantly decreased oxidative stress by decreasing malondialdehyde and nitric oxide tissue levels with a significant increase in spectrophotometric measurements of superoxide dismutase, catalase, and reduced glutathione levels. DPGZ treatment resulted in a significant anti-inflammatory effect as indicated by suppression in myeloperoxidase activity, MCP-1, IL-1ß, IL-18, and TNF-α levels. DPGZ treatment also increased p-AMPK/t-AMPK with a significant reduction in NF-kB P65 binding activity and NFĸB p65 (pSer536) levels. These effects of DPGZ were accompanied by a significant reduction in NLRP3 levels and NLRP3 gene expression and a significant decrease in caspase-1 activity, which were also confirmed by histopathological examinations. We conclude that DPGZ antioxidant and anti-inflammatory activity may occur through regulation of AMPK/NFĸB pathway and inhibition of NLRP3 activation. These results suggest that DPGZ represents a promising intervention for the treatment of ALI, particularly in patients with type 2 diabetes.


Subject(s)
Acute Lung Injury/prevention & control , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Inflammation/prevention & control , Signal Transduction/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , AMP-Activated Protein Kinases/metabolism , Acute Lung Injury/chemically induced , Animals , Benzhydryl Compounds/chemistry , Disease Models, Animal , Glucosides/chemistry , Inflammation/chemically induced , Lipopolysaccharides , Male , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 2 Inhibitors/chemistry
7.
Biomed Pharmacother ; 145: 112455, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34844106

ABSTRACT

Hepatocellular carcinoma (HCC) is on the rise worldwide, and its incidence in diabetic patients is two to three times that of non-diabetics. Current therapeutic options fail to provide considerable survival benefits to patients with HCC. There is a strong possibility that the FDA-approved antidiabetic combination of empagliflozin and metformin could show complementary effects to control HCC progression. However, their multitarget effects have not yet been studied on HCC development. Therefore, the present study aims to evaluate the antitumorigenic activity of this combination in non-diabetic mice with diethylnitrosamine-induced HCC. Empagliflozin/metformin combination prolonged survival and improved histological features of mice livers. Additionally, Empagliflozin/metformin showed anti-inflammatory potential and relieved oxidative stress. On the one hand these effects are likely attributed to the ability of metformin to inactivate NF-κB in an AMPK-dependent mechanism and on the other hand to the ability of the empagliflozin to inhibit the MAPKs, p38 and ERK1/2. Empagliflozin also showed a less robust effect on AMPK than that of metformin. Moreover, empagliflozin enhanced the autophagy inducing activity of metformin. Furthermore, empagliflozin/metformin exhibited increased apoptotic potential. Consequently, empagliflozin augmented the antitumorigenic function of metformin by exerting better control of angiogenesis, and metastasis. To conclude, our findings suggest empagliflozin as an ideal adjunct to metformin for the inhibition of HCC progression. In addition, since the incidence of hypoglycemia is minimal due to insulin-independent mechanism of action of both treatments, empagliflozin/metformin could be a promising therapeutic modality for the management of diabetic patients with HCC; and even non diabetic ones.


Subject(s)
Benzhydryl Compounds/pharmacology , Carcinoma, Hepatocellular , Glucosides/pharmacology , Liver Neoplasms , Metformin/pharmacology , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Disease Progression , Hypoglycemic Agents/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Kinase Kinases/metabolism , Mice , NF-kappa B/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
8.
Toxicology ; 463: 152994, 2021 11.
Article in English | MEDLINE | ID: mdl-34678320

ABSTRACT

Overexposure to carcinogenic precursor, benzo[a]pyrene [BaP], modulates the lung immune microenvironment. The present review seeks to elucidate novel pathways behind the tumor effect of BaP in the lungs, emphasizing immunomodulatory mediators and immune cells. In this review, BaP reprograms lung immune microenvironment through modulating transforming growth factor-beta (TGF-ß), programmed cell death 1 (PD-1), cytotoxic T lymphocyte antigen-4 (CTLA-4), Interleukin 12 (IL-12), indoleamine 2,3 dioxygenase (IDO), forkhead box protein P3 (FOXP3) and interferon-gamma (IFN-γ) levels. Moreover, BaP modulated lung immune cellular architecture such as dendritic cells, T cells, Tregs, macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs). All mentioned changes in immune architecture and mediators lead to the induction of lung cancer.


Subject(s)
Benzo(a)pyrene/toxicity , Lung Neoplasms/chemically induced , Lung/drug effects , Animals , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogens/toxicity , Humans , Lung/immunology , Lung/pathology , Lung Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
9.
Life Sci ; 286: 120070, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34688695

ABSTRACT

AIM: Metformin and empagliflozin combined therapy may have complementary effects that go beyond the well-recognized targets of their monotherapy through AMPK activation. Therefore, the current study was designed to investigate for the first time the hepatoprotective effects of such combination therapy in the carbon tetrachloride (CCl4)-induced hepatic fibrosis model in mice. MATERIALS AND METHODS: Determination of liver enzymes and the liver content of oxidative stress parameters, and hydroxyproline were performed biochemically. ELISA was performed to measure PDGF-BB, TNF-α, TGF-ß, TIMP-1, AMPK, p-mTOR, NF-κB P65 binding activity, p38 MAPKα, JNK1/2 and ERK1/2. Real-time qPCR was conducted to determine Col1a1 and α-SMA. In addition, histopathological examination using H&E and Masson's trichrome stain were performed for determination of histopathological changes. KEY FINDINGS: Empagliflozin inhibited the activation of p38 MAPK and ERK1/2 and exhibited a weak AMPKα stimulation. On the other hand, metformin exerted a more robust stimulatory action on the AMPKα that was accompanied by a notable decrease in the NF-κB nuclear binding activity and a decline in the p-mTOR levels. Nevertheless, the effect of metformin on MAPK kinases was insignificant. Our results revealed that blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhanced the antifibrotic effect of metformin and augmented its AMPK-induced NF-κB inactivation. SIGNIFICANCE: As diabetes is one of the most common risk factors for liver fibrosis, the use of antidiabetic drugs is expected to improve therapeutic outcome. Therefore, metformin/empagliflozin combined therapy could be promising in preventing hepatic inflammation and fibrosis via exhibiting complementary effects particularly in diabetic patients.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Liver Cirrhosis/drug therapy , Metformin/pharmacology , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Animals , Benzhydryl Compounds/metabolism , Carbon Tetrachloride/pharmacology , Drug Therapy, Combination/methods , Female , Glucosides/metabolism , Hepatocytes/metabolism , Liver/metabolism , Liver Cirrhosis/physiopathology , MAP Kinase Signaling System/physiology , Male , Metformin/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Primary Cell Culture , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Front Pharmacol ; 12: 719984, 2021.
Article in English | MEDLINE | ID: mdl-34489707

ABSTRACT

Empagliflozin and metformin are widely used for the treatment of type 2 diabetes. These drugs showed marked anti-inflammatory effects in different animal models via enhancing AMPK activity. Yet, the protective anti-inflammatory effects of their combination against ulcerative colitis have not been previously investigated. The current study aimed to explore the potential of empagliflozin/metformin combination to mitigate the DSS-induced rat colitis model. The modulating effects of empagliflozin and metformin on the AMPK/mTOR/NLRP3 axis and T cell polarization were delineated. In this study, distal colons were examined for macroscopic and microscopic pathological alterations. ELISA, qRT-PCR, and immunohistochemistry techniques were applied to detect proteins and cytokines involved in AMPK/mTOR/NLRP3 axis and T Cell polarization. Oral administration of empagliflozin (10 mg/kg/day) and metformin (200 mg/kg/day) combination alleviated colitis as revealed by the reduced disease activity index, macroscopic damage index, colon weight/length ratio, and histopathologic scoring values. Interestingly, empagliflozin/metformin combination significantly enhanced AMPK phosphorylation and depressed mTOR and NLRP3 expression leading to a subsequent reduction in caspase-1 cleavage and inhibition of several inflammatory cytokines, including IL-1ß, and IL-18. Reduced mTOR expression and reduced IL-6 levels led to a reduction in Th17 cell polarization and maintenance. Together, the current study reveals that the protective effects of empagliflozin and metformin against DSS-induced colitis are fundamentally mediated via enhancing AMPK phosphorylation. Since adult humans with diabetes mellitus are at greater risk for developing inflammatory bowel diseases, clinical application of empagliflozin/metformin combination represents a novel therapeutic approach for treating diabetic patients with ulcerative colitis.

11.
Pharmaceuticals (Basel) ; 14(4)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917884

ABSTRACT

Inflammasome targeting and controlling dysbiosis are promising therapeutic approaches to control ulcerative colitis. This report is the first to investigate the mechanisms underlying the coloprotective effects of rosuvastatin and Lactobacillus and their combined therapy on dextran sodium sulfate (DSS)-induced colitis in high-fat diet (HFD)-fed rats. Our results demonstrate the aggravation of intestinal inflammation as a consequence of an HFD following DSS administration. An association between dyslipidemia, LDL oxidation, CD36 expression, ROS generation, thioredoxin-interacting protein (TXNIP) upregulation, and NLRP3 inflammasome activation was demonstrated by DSS exposure in HFD-fed rats. We demonstrated that rosuvastatin/Lactobacillus significantly suppressed the DSS/HFD-induced increase in colon weight/length ratio, DAI, MDI, and myeloperoxidase, as well as corrected dysbiosis and improved histological characteristics. Additionally, caspase-1 activity and IL-1ß-driven pyroptotic activity was significantly reduced. Rosuvastatin/Lactobacillus showed prominent anti-inflammatory effects as revealed by the IL-10/IL-12 ratio and the levels of TNF-α and IL-6. These latter effects may be attributed to the inhibition of phosphorylation-induced activation of NF-κB and a concomitant reduction in the expression of NLRP3, pro-IL-1ß, and pro-IL-18. Furthermore, rosuvastatin/Lactobacillus reduced Ox-LDL-induced TXNIP and attenuated the inflammatory response by inhibiting NLRP3 inflammasome assembly. To conclude, rosuvastatin/Lactobacillus offers a safe and effective strategy for the management of ulcerative colitis.

12.
Front Pharmacol ; 12: 720173, 2021.
Article in English | MEDLINE | ID: mdl-35095479

ABSTRACT

HIF-1α is a key factor promoting the development of hepatocellular carcinoma (HCC). As well, AKT-AMPKα-mTOR signaling is a promising target for cancer therapy. Yet, the AKT-AMPKα-mTOR-dependent activation of HIF-1α has not been studied in livers with HCC. In addition, the mechanisms underlying the potential antineoplastic effects of sitagliptin (STGPT), an antidiabetic agent, have not yet been elucidated. For that purpose, the N-nitrosodiethylamine (NDEA)-induced HCC mouse model was used in the present study using a dose of 100 mg/kg/week, i.p., for 8 weeks. NDEA-induced HCC mice received STGPT 20, 40, or 80 mg/kg starting on day 61 up to day 120. The present study revealed that STGPT inhibited HIF-1α activation via the interference with the AKT-AMPKα-mTOR axis and the interruption of IKKß, P38α, and ERK1/2 signals as well. Accordingly, STGPT prolonged the survival, restored the histological features and improved liver function. Additionally, STGPT inhibited angiogenesis, as revealed by a significant downregulation in the VEGF and mRNA expression of CD309 with concomitant inhibition of tissue invasion was evident by an increased ratio of TIMP-1/MMP-2. STGPT exhibited apoptotic stimulatory effect as indicated upon calculating the BCL-2/Bax ratio and by the gene expression of p53. The decrease in AFP and liver index calculation, gene expression of Ki-67 confirmed the antiproliferative activity of STGPT. The anti-inflammatory potential was revealed by the decreased TNF-α level and the downregulation of MCP-1 gene expression. Moreover, an antifibrotic potential was supported by lower levels of TGF-ß. These effects appear to be GLP1R-independent. The present study provides a potential basis for repurposing STGPT for the inhibition of HCC progression. Since STGPT is unlikely to cause hypoglycemia, it may be promising as monotherapy or adjuvant therapy to treat diabetic or even normoglycemic patients with HCC.

13.
Chem Biol Interact ; 333: 109330, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33245929

ABSTRACT

AIM: Benzo[a]pyrene [BP] is one of the major carcinogenic precursors of cigarette smoke that primary affects the lung at its first proximity. The goal of the current research was to elucidate new mechanisms underlying the tumorigenic impact of oral BP in the lung of mice, with focus on immunosuppressive effects and cancer stemming properties. METHODS: Female albino mice (n = 44) were divided into 2 groups: normal control and BP group. BP was administered orally to mice (50 mg/kg body weight), twice a week for four weeks in succession. At the end of experiment (22 weeks), gene expression were measured for transforming growth factor-ß (TGF-ß), cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed death ligand 1(PD-L1), forkhead box protein P3 (FOXP3) and interleukin 12 (IL-12) and CD83+, CD8+ and CD166+ cell percentage were measured in lung tissue. RESULTS: The results indicated the tumorigenic role of BP in the lung which was evidenced by histopathological examination. BP group also showed immunosuppressive role which evidenced by increased expression of lung TGF-ß, CTLA-4, PD-L1, FOXP3 genes and decreased expression of lung IL-12 gene compared with normal control group. BP group also showed decreased CD83+ cells, CD8+ cells and increased number of CD166+ cells. CONCLUSION: Our findings indicated that BP has immunosuppressive role in lung cancer besides increasing the percentage of cancer stem like cells.


Subject(s)
Benzo(a)pyrene/pharmacology , Carcinogenesis/drug effects , Immunosuppressive Agents/pharmacology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Animals , Antigens, CD/metabolism , B7-H1 Antigen/genetics , CTLA-4 Antigen/genetics , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-12/genetics , Mice , Survival Analysis , Transforming Growth Factor beta/genetics , Tumor Burden/drug effects
14.
Clin Exp Med ; 17(2): 185-191, 2017 May.
Article in English | MEDLINE | ID: mdl-27085325

ABSTRACT

Although sorafenib was approved as antiangiogenic agent in case of hepatocellular carcinoma (HCC), the pathways mediating its antitumorigenic effects were not fully examined in vivo. This study was conducted to elucidate the molecular mechanisms underlying the antineoplastic effect of sorafenib in livers of rats exposed to the hepatocarcinogen diethyl nitrosamine (DENA) regarding oxidative stress, proliferation, and apoptotic pathways. Male albino rats were divided into three groups: normal control, DENA group, and sorafenib group. Sorafenib (10 mg/kg) was given daily to rats orally for 2 weeks, started 6 weeks after DENA (200 mg/kg, single i.p. dose). The histopathological results proved that sorafenib corrected neoplastic changes in the liver as evidenced by a decrease in size of hepatocellular foci. The liver index, glutathione, as well as Bcl-2 were significantly decreased in sorafenib group compared with DENA group. Sorafenib also exhibited antiproliferative effect through suppression of gene expression of cyclin D1 and ß-catenin. Thus, the apoptotic and proliferative pathways in HCC could be interrupted by sorafenib, supporting the role of sorafenib as antineoplastic agent and nominating it as a candidate drug for other neoplasms.


Subject(s)
Antineoplastic Agents/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Cyclin D1/analysis , Disease Models, Animal , Glutathione/analysis , Histocytochemistry , Liver/pathology , Liver Neoplasms/chemically induced , Male , Niacinamide/administration & dosage , Niacinamide/pharmacology , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-bcl-2/analysis , Rats , Sorafenib , Treatment Outcome , beta Catenin/analysis
15.
Nutr Cancer ; 68(6): 1044-51, 2016.
Article in English | MEDLINE | ID: mdl-27383944

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer-related mortality worldwide. The current work was designed to elucidate the molecular mechanisms underlying the antitumorigenic effect of pomegranate hull extract (PHE) in livers of rats exposed to the hepatocarcinogen diethyl nitrosamine (DENA) with emphasis on oxidative stress, proliferation, and apoptosis. Male albino rats were divided into three groups: normal control, DENA group, and PHE group. PHE was given to rats orally 3 times weekly for 10 wk, 4 wk before and 6 wk after DENA (200 mg/kg, single i.p. dose). The results indicated a prophylactic effect of PHE against neoplastic changes in the liver, which was evidenced by the decrease of tumor size, liver index, and the anti-apoptotic protein Bcl-2; and the increase of glutathione. PHE group also showed decreased expression of liver cyclin D1 and ß-catenin genes compared with DENA group. It is proved that PHE has antitumorigenic effect and could be a candidate for anticancer drugs.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Hepatocellular/prevention & control , Dietary Supplements , Liver Neoplasms/prevention & control , Liver/metabolism , Lythraceae/chemistry , Plant Extracts/therapeutic use , Animals , Antioxidants/therapeutic use , Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/diet therapy , Carcinoma, Hepatocellular/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Diethylnitrosamine/toxicity , Fruit/chemistry , Fruit/economics , Gene Expression Regulation, Neoplastic/drug effects , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/diet therapy , Liver Neoplasms/pathology , Male , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , Rats , Survival Rate , Tumor Burden/drug effects , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...