1
|
Abbasloo E, Amiresmaili S, Shirazpour S, Khaksari M, Kobeissy F, Thomas TC. Satureja khuzistanica Jamzad essential oil and pure carvacrol attenuate TBI-induced inflammation and apoptosis via NF-κB and caspase-3 regulation in the male rat brain. Sci Rep 2023; 13:4780. [PMID: 36959464 PMCID: PMC10036533 DOI: 10.1038/s41598-023-31891-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/20/2023] [Indexed: 03/25/2023] Open
Abstract
Traumatic brain injury (TBI) causes progressive dysfunction that induces biochemical and metabolic changes that lead to cell death. Nevertheless, there is no definitive FDA-approved therapy for TBI treatment. Our previous immunohistochemical results indicated that the cost-effective natural Iranian medicine, Satureja khuzistanica Jamzad essential oil (SKEO), which consists of 94.16% carvacrol (CAR), has beneficial effects such as reducing neuronal death and inflammatory markers, as well as activating astrocytes and improving neurological outcomes. However, the molecular mechanisms of these neuroprotective effects have not yet been elucidated. This study investigated the possible mechanisms involved in the anti-inflammatory and anti-apoptotic properties of SKEO and CAR after TBI induction. Eighty-four male Wistar rats were randomly divided into six groups: Sham, TBI, TBI + Vehicle, TBI + CAR (100 and 200 mg/kg), and TBI + SKEO (200 mg/kg) groups. After establishing the "Marmarou" weight drop model, diffuse TBI was induced in the rat brain. Thirty minutes after TBI induction, SKEO & CAR were intraperitoneally injected. One day after TBI, injured rats exhibited significant brain edema, neurobehavioral dysfunctions, and neuronal apoptosis. Western blot results revealed upregulation of the levels of cleaved caspase-3, NFκB p65, and Bax/Bcl-2 ratio, which was attenuated by CAR and SKEO (200 mg/kg). Furthermore, the ELISA results showed that CAR treatment markedly prevents the overproduction of the brain pro-inflammatory cytokines, including IL-1β, TNF-α, and IL-6. Moreover, the neuron-specific enolase (NSE) immunohistochemistry results revealed the protective effect of CAR and SKEO on post-TBI neuronal death. The current study revealed that the possible neuroprotective mechanisms of SKEO and CAR might be related to (at least in part) modulating NF-κB regulated inflammation and caspase-3 protein expression. It also suggested that CAR exerts more potent protective effects than SKEO against TBI. Nevertheless, the administration of SKEO and CAR may express a novel therapeutic approach to ameliorate TBI-related secondary phase neuropathological outcomes.
Collapse
Affiliation(s)
- Elham Abbasloo
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Sara Shirazpour
- Department of Physiology and Pharmacology, Faculty of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Neurotrauma, Multiomics and Biomarkers, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, USA
- Translational Neurotrauma Research Program, Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, USA
| |
Collapse
|
2
|
Khaligh SF, Asoodeh A. Recent advances in the bio-application of microalgae-derived biochemical metabolites and development trends of photobioreactor-based culture systems. 3 Biotech 2022; 12:260. [PMID: 36072963 PMCID: PMC9441132 DOI: 10.1007/s13205-022-03327-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022] Open
Abstract
Microalgae are microscopic algae in sizes ranging from a few micrometers to several hundred micrometers. On average, half of the oxygen in the atmosphere is produced by the photosynthetic process of microalgae, so the role of these microorganisms in the life cycle of the planet is very significant. Pharmaceutical products derived from microalgae and commercial developments of a variety of supplements extracted from them originate from a variety of their specific secondary metabolites. Many of these microalgae are a reservoir of unique biological compounds including carotenoids, antioxidants, fatty acids, polysaccharides, enzymes, polymers, peptides, pigments, toxins and sterols with antimicrobial, antiviral, antifungal, antiparasitic, anticoagulant, and anticancer properties. The present work begins with an introduction of the importance of microalgae in renewable fuels and biodiesel production, the development of healthy food industry, and the creation of optimal conditions for efficient biomass yield. This paper provides the latest research related to microalgae-derived substances in the field of improving drug delivery, immunomodulatory, and anticancer attributes. Also, the latest advances in algal biocompounds to combat the COVID-19 pandemic are presented. In the subject of cultivation and growth of microalgae, the characteristics of different types of photobioreactors, especially their latest forms, are fully discussed along with their advantages and obstacles. Finally, the potential of microalgae biomass in biotechnological applications, biofuel production, as well as various biomass harvesting methods are described.
Collapse
Affiliation(s)
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
3
|
Kang N, Xie X, Zhou X, Wang Y, Chen S, Qi R, Liu T, Jiang H. Identification and validation of EMT-immune-related prognostic biomarkers CDKN2A, CMTM8 and ILK in colon cancer. BMC Gastroenterol 2022; 22:190. [PMID: 35429970 PMCID: PMC9013447 DOI: 10.1186/s12876-022-02257-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/31/2022] [Indexed: 12/26/2022] Open
Abstract
Colon cancer (CC) is a disease with high incidence and mortality rate. The interaction between epithelial-mesenchymal transition (EMT) and immune status has important clinical significance. We aim to identify EMT-immune-related prognostic biomarkers in colon cancer. The GEO2R and GEPIA 2.0 were utilized to calculate the differential expression genes between CC and normal mucosa. Immport, InnateDB and EMTome databases were used to define EMT-immune-related genes. We conducted batch prognostic analysis by TCGA data. The expression patterns were verified by multiple datasets and lab experiments. GEPIA 2.0 and TIMER 2.0 were utilized to analyze the correlation of the hub genes with EMT markers and immune infiltration. GeneMANIA, STRING, and Metascape were used for co-expression and pathway enrichment analysis. Finally, we established a signature by the method of multivariate Cox regression analysis. CDKN2A, CMTM8 and ILK were filtered out as prognostic genes. CDKN2A and CMTM8 were up-regulated, while ILK was down-regulated in CC. CDKN2A was positively correlated with infiltration of macrophages, Th2 cells, Treg cells, and negatively correlated with NK cells. CMTM8 was negatively correlated with CD8+ T cells, dendritic cells, and NK cells. ILK was positively correlated with CD8+ T cells and dendritic cells. Moreover, CDKN2A, CMTM8 and ILK were significantly correlated with EMT markers. The three genes could participate in the TGF-β pathway. The prognosis model established by the three hub genes was an independent prognosis factor, which can better predict the prognosis. CDKN2A, CMTM8 and ILK are promising prognostic biomarkers and may be potential therapeutic targets in colon cancer.
Collapse
|
4
|
Alanazi MM, Eissa IH, Alsaif NA, Obaidullah AJ, Alanazi WA, Alasmari AF, Albassam H, Elkady H, Elwan A. Design, synthesis, docking, ADMET studies, and anticancer evaluation of new 3-methylquinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers. J Enzyme Inhib Med Chem 2021; 36:1760-1782. [PMID: 34340610 PMCID: PMC8344243 DOI: 10.1080/14756366.2021.1956488] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR-2) plays a critical role in cancer angiogenesis. Inhibition of VEGFR-2 activity proved effective suppression of tumour propagation. Accordingly, two series of new 3-methylquinoxaline derivatives have been designed and synthesised as VEGFR-2 inhibitors. The synthesised derivatives were evaluated in vitro for their cytotoxic activities against MCF-7and HepG2 cell lines. In addition, the VEGFR-2 inhibitory activities of the target compounds were estimated to indicate the potential mechanism of their cytotoxicity. To a great extent, the results of VEGFR-2 inhibition were highly correlated with that of cytotoxicity. Compound 27a was the most potent VEGFR-2 inhibitor with IC50 of 3.2 nM very close to positive control sorafenib (IC50 = 3.12 nM). Such compound exhibited a strong cytotoxic effect against MCF-7 and HepG2, respectively with IC50 of 7.7 and 4.5 µM in comparison to sorafenib (IC50 = 3.51 and 2.17 µM). In addition, compounds 28, 30f, 30i, and 31b exhibited excellent VEGFR-2 inhibition activities (IC50 range from 4.2 to 6.1 nM) with promising cytotoxic activity. Cell cycle progression and apoptosis induction were investigated for the most active member 27a. Also, the effect of 27a on the level of caspase-3, caspase-9, and BAX/Bcl-2 ratio was determined. Molecular docking studies were implemented to interpret the binding mode of the target compounds with the VEGFR-2 pocket. Furthermore, toxicity and ADMET calculations were performed for the synthesised compounds to study their pharmacokinetic profiles.
Collapse
Affiliation(s)
- Mohammed M. Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Nawaf A. Alsaif
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad J. Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wael A. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
5
|
Liu C, Seeram NP, Ma H. Small molecule inhibitors against PD-1/PD-L1 immune checkpoints and current methodologies for their development: a review. Cancer Cell Int 2021; 21:239. [PMID: 33906641 PMCID: PMC8077906 DOI: 10.1186/s12935-021-01946-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Programmed death-1/programmed death ligand-1 (PD-1/PD-L1) based immunotherapy is a revolutionary cancer therapy with great clinical success. The majority of clinically used PD-1/PD-L1 inhibitors are monoclonal antibodies but their applications are limited due to their poor oral bioavailability and immune-related adverse effects (irAEs). In contrast, several small molecule inhibitors against PD-1/PD-L1 immune checkpoints show promising blockage effects on PD-1/PD-L1 interactions without irAEs. However, proper analytical methods and bioassays are required to effectively screen small molecule derived PD-1/PD-L1 inhibitors. Herein, we summarize the biophysical and biochemical assays currently employed for the measurements of binding capacities, molecular interactions, and blocking effects of small molecule inhibitors on PD-1/PD-L1. In addition, the discovery of natural products based PD-1/PD-L1 antagonists utilizing these screening assays are reviewed. Potential pitfalls for obtaining false leading compounds as PD-1/PD-L1 inhibitors by using certain binding bioassays are also discussed in this review.
Collapse
Affiliation(s)
- Chang Liu
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| | - Navindra P Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Avedisian Hall Lab 440, 7 Greenhouse Road, Kingston, RI, 02881, USA.
| |
Collapse
|
6
|
He L, Shi X, Chen R, Wu Z, Yang Z, Li Z. Association of Mental Health-Related Proteins DAXX, DRD3, and DISC1 With the Progression and Prognosis of Chondrosarcoma. Front Mol Biosci 2019; 6:134. [PMID: 31850367 PMCID: PMC6888811 DOI: 10.3389/fmolb.2019.00134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Chondrosarcoma is the second most common malignant bone tumor. Current therapies remain ineffective, resulting in poor prognoses. Biomarkers for chondrosarcoma and predictors of its prognosis have not been established. Mental health-related proteins have been associated with the pathogenesis, progression, and prognosis of many cancers, but their association with chondrosarcoma has not been reported. In this study, the expression and clinicopathological significance of the mental health-related proteins DAXX, DRD3, and DISC1 in chondrosarcoma tissue samples were examined, over an 84-months follow-up period. In immunohistochemical analysis, the rates of positive DAXX, DRD3, and DISC1 expression were significantly higher in chondrosarcoma than in osteochondroma tissue (P < 0.01). The percentages of positive DAXX, DRD3, and DISC1 expression were significantly lower in tissues with good differentiation (P < 0.01), AJCC stage I/ II (P < 0.01), Enneking stage I (P < 0.01), and non-metastasis (P < 0.05), respectively. In Kaplan-Meier survival analysis, significantly shorter mean survival times were associated with moderate and poor differentiation (P = 0.000), AJCC stage III/IV (P = 0.000), Enneking stage II/III (P = 0.000), metastasis (P = 0.019), invasion (P = 0.013), and positive DAXX (P = 0.012), and/or DRD3 (P = 0.018) expression. In Cox regression analysis, moderate and poor differentiation (P = 0.006), AJCC stage III/IV (P = 0.013), Enneking stage II/III (P = 0.016), metastasis (P = 0.033), invasion (P = 0.011), and positive DAXX (P = 0.033), and/or DRD3 (P = 0.025) staining correlated negatively with the postoperative survival rate and positively with mortality. In competing-risks regression analysis, differentiation (P = 0.005), metastasis (P = 0.014), invasion (P = 0.028), AJCC stage (P = 0.003), Enneking stage (P = 0.036), and DAXX (P = 0.039), and DRD3(P = 0.019) expression were independent predictors of death from chondrosarcoma. The areas under receiver operating characteristic curves for DAXX, DRD3, and DISC1 expression were 0.673 (95% CI, 0.557-0.788; P = 0.010), 0.670 (95% CI, 0.556-0.784; P = 0.011), and 0.688 (95% CI, 0.573-0.802; P = 0.005), respectively. These results suggest that DAXX, DRD3, and DISC1 could serve as biomarkers of chondrosarcoma progression and predictors of its prognosis.
Collapse
Affiliation(s)
- Lile He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| | - Xiangyu Shi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruiqi Chen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| | - Zhengchun Wu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhulin Yang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| |
Collapse
|
7
|
PGAM5-CypD pathway is involved in bromocriptine-induced RIP3/MLKL-dependent necroptosis of prolactinoma cells. Biomed Pharmacother 2019; 111:638-648. [DOI: 10.1016/j.biopha.2018.12.128] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/27/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022] Open
|
8
|
Zemková H, Stojilkovic SS. Neurotransmitter receptors as signaling platforms in anterior pituitary cells. Mol Cell Endocrinol 2018; 463:49-64. [PMID: 28684290 PMCID: PMC5752632 DOI: 10.1016/j.mce.2017.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/29/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
Abstract
The functions of anterior pituitary cells are controlled by two major groups of hypothalamic and intrapituitary ligands: one exclusively acts on G protein-coupled receptors and the other activates both G protein-coupled receptors and ligand-gated receptor channels. The second group of ligands operates as neurotransmitters in neuronal cells and their receptors are termed as neurotransmitter receptors. Most information about pituitary neurotransmitter receptors was obtained from secretory studies, RT-PCR analyses of mRNA expression and immunohistochemical and biochemical analyses, all of which were performed using a mixed population of pituitary cells. However, recent electrophysiological and imaging experiments have characterized γ-aminobutyric acid-, acetylcholine-, and ATP-activated receptors and channels in single pituitary cell types, expanding this picture and revealing surprising differences in their expression between subtypes of secretory cells and between native and immortalized pituitary cells. The main focus of this review is on the electrophysiological and pharmacological properties of these receptors and their roles in calcium signaling and calcium-controlled hormone secretion.
Collapse
Affiliation(s)
- Hana Zemková
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology, ASCR, Prague, Czech Republic.
| | - Stanko S Stojilkovic
- Sections on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
9
|
Kerdudo A, Ellong EN, Burger P, Gonnot V, Boyer L, Chandre F, Adenet S, Rochefort K, Michel T, Fernandez X. Chemical Composition, Antimicrobial and Insecticidal Activities of Flowers Essential Oils ofAlpinia zerumbet(Pers.) B.L.Burtt& R.M.Sm. from Martinique Island. Chem Biodivers 2017; 14. [DOI: 10.1002/cbdv.201600344] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/09/2016] [Indexed: 12/25/2022]
Affiliation(s)
| | - Emy Njoh Ellong
- Pôle Agroalimentaire Régional de Martinique (P.A.R.M.); Impasse Petit Morne No. 375 97232 Lamentin, Martinique France
| | | | | | - Laurent Boyer
- INSERM, U1065; Centre Méditerranéen de Médecine Moléculaire, C3M; Toxines Microbiennes dans la relation hôte-pathogène; Nice France
- UFR Médecine; Université de Nice-Sophia-Antipolis; Nice France
| | - Fabrice Chandre
- UMR Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (IRD 224-CNRS 5290-UM); Institut de Recherche pour le Développement (IRD); 911 Avenue Agropolis 34394 Montpellier Cedex 5 France
| | - Sandra Adenet
- Pôle Agroalimentaire Régional de Martinique (P.A.R.M.); Impasse Petit Morne No. 375 97232 Lamentin, Martinique France
| | - Katia Rochefort
- Pôle Agroalimentaire Régional de Martinique (P.A.R.M.); Impasse Petit Morne No. 375 97232 Lamentin, Martinique France
| | | | | |
Collapse
|
10
|
Usman M, Arjmand F, Khan RA, Alsalme A, Ahmad M, Tabassum S. Biological evaluation of dinuclear copper complex/dichloroacetic acid cocrystal against human breast cancer: design, synthesis, characterization, DFT studies and cytotoxicity assays. RSC Adv 2017; 7:47920-47932. [DOI: 10.1039/c7ra08262b] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Binuclear copper(ii) cocrystal “[Cu2(valdien)2⋯2Cl2CHCOOH],” 1 was synthesized from H2valdien scaffold and anticancer drug pharmacophore “dichloroacetic acid” embedded with two Cu(ii) connected via a hydrogen bonded network.
Collapse
Affiliation(s)
- Mohammad Usman
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Farukh Arjmand
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Rais Ahmad Khan
- Department of Chemistry
- College of Science
- King Saud University
- Riyadh 11451
- Saudi Arabia
| | - Ali Alsalme
- Department of Chemistry
- College of Science
- King Saud University
- Riyadh 11451
- Saudi Arabia
| | - Musheer Ahmad
- Department of Applied Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Sartaj Tabassum
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| |
Collapse
|
11
|
Sathe G, Pinto SM, Syed N, Nanjappa V, Solanki HS, Renuse S, Chavan S, Khan AA, Patil AH, Nirujogi RS, Nair B, Mathur PP, Prasad TSK, Gowda H, Chatterjee A. Phosphotyrosine profiling of curcumin-induced signaling. Clin Proteomics 2016; 13:13. [PMID: 27307780 PMCID: PMC4908701 DOI: 10.1186/s12014-016-9114-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Curcumin, derived from the rhizome Curcuma longa, is a natural anti-cancer agent and has been shown to inhibit proliferation and survival of tumor cells. Although the anti-cancer effects of curcumin are well established, detailed understanding of the signaling pathways altered by curcumin is still lacking. In this study, we carried out SILAC-based quantitative proteomic analysis of a HNSCC cell line (CAL 27) to investigate tyrosine signaling in response to curcumin. RESULTS Using high resolution Orbitrap Fusion Tribrid Fourier transform mass spectrometer, we identified 627 phosphotyrosine sites mapping to 359 proteins. We observed alterations in the level of phosphorylation of 304 sites corresponding to 197 proteins upon curcumin treatment. We report here for the first time, curcumin-induced alterations in the phosphorylation of several kinases including TNK2, FRK, AXL, MAPK12 and phosphatases such as PTPN6, PTPRK, and INPPL1 among others. Pathway analysis revealed that the proteins differentially phosphorylated in response to curcumin are known to be involved in focal adhesion kinase signaling and actin cytoskeleton reorganization. CONCLUSIONS The study indicates that curcumin may regulate cellular processes such as proliferation and migration through perturbation of the focal adhesion kinase pathway. This is the first quantitative phosphoproteomics-based study demonstrating the signaling events that are altered in response to curcumin. Considering the importance of curcumin as an anti-cancer agent, this study will significantly improve the current knowledge of curcumin-mediated signaling in cancer.
Collapse
Affiliation(s)
- Gajanan Sathe
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Manipal University, Madhav Nagar, Manipal, 576104 India
| | - Sneha M Pinto
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, 575018 India
| | - Nazia Syed
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605014 India
| | - Vishalakshi Nanjappa
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Amrita School of Biotechnology, Amrita University, Kollam, 690525 India
| | - Hitendra S Solanki
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | - Santosh Renuse
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Amrita School of Biotechnology, Amrita University, Kollam, 690525 India
| | - Sandip Chavan
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Manipal University, Madhav Nagar, Manipal, 576104 India
| | - Aafaque Ahmad Khan
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | - Arun H Patil
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | - Raja Sekhar Nirujogi
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,Centre of Excellence in Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, 605014 India
| | - Bipin Nair
- Amrita School of Biotechnology, Amrita University, Kollam, 690525 India
| | | | - T S Keshava Prasad
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, 575018 India.,NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| | - Harsha Gowda
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, 575018 India
| | - Aditi Chatterjee
- Institute of Bioinformatics, Unit I, 7th Floor, Discoverer Building, International Tech Park, Bangalore, 560066 India.,YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore, 575018 India
| |
Collapse
|
12
|
Fatima A, Abdul ABH, Abdullah R, Karjiban RA, Lee VS. Binding mode analysis of zerumbone to key signal proteins in the tumor necrosis factor pathway. Int J Mol Sci 2015; 16:2747-66. [PMID: 25629232 PMCID: PMC4346863 DOI: 10.3390/ijms16022747] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/07/2015] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the second most common cancer among women worldwide. Several signaling pathways have been implicated as causative and progression agents. The tumor necrosis factor (TNF) α protein plays a dual role in promoting and inhibiting cancer depending largely on the pathway initiated by the binding of the protein to its receptor. Zerumbone, an active constituent of Zingiber zerumbet, Smith, is known to act on the tumor necrosis factor pathway upregulating tumour necrosis factor related apoptosis inducing ligand (TRAIL) death receptors and inducing apoptosis in cancer cells. Zerumbone is a sesquiterpene that is able to penetrate into the hydrophobic pockets of proteins to exert its inhibiting activity with several proteins. We found a good binding with the tumor necrosis factor, kinase κB (IKKβ) and the Nuclear factor κB (NF-κB) component proteins along the TNF pathway. Our results suggest that zerumbone can exert its apoptotic activities by inhibiting the cytoplasmic proteins. It inhibits the IKKβ kinase that activates the NF-κB and also binds to the NF-κB complex in the TNF pathway. Blocking both proteins can lead to inhibition of cell proliferating proteins to be downregulated and possibly ultimate induction of apoptosis.
Collapse
Affiliation(s)
- Ayesha Fatima
- UPM-MAKNA Cancer Research Laboratory, Institute of Biosciences, University Putra Malaysia, 43400 Serdang, Malaysia.
| | - Ahmad Bustamam Hj Abdul
- UPM-MAKNA Cancer Research Laboratory, Institute of Biosciences, University Putra Malaysia, 43400 Serdang, Malaysia.
| | - Rasedee Abdullah
- UPM-MAKNA Cancer Research Laboratory, Institute of Biosciences, University Putra Malaysia, 43400 Serdang, Malaysia.
| | - Roghayeh Abedi Karjiban
- Department of Chemistry, Faculty of Science, University Putra Malaysia, 43400 Serdang, Malaysia.
| | - Vannajan Sanghiran Lee
- Department of Chemistry, Faculty of Science, University Malaya, Petaling Jaya, 50603 Selangor, Malaysia.
| |
Collapse
|
13
|
Pornour M, Ahangari G, Hejazi SH, Ahmadkhaniha HR, Akbari ME. Dopamine Receptor Gene (DRD1-DRD5) Expression Changes as Stress Factors Associated with Breast Cancer. Asian Pac J Cancer Prev 2015; 15:10339-43. [DOI: 10.7314/apjcp.2014.15.23.10339] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
14
|
Wehinger S, Ortiz R, Díaz MI, Aguirre A, Valenzuela M, Llanos P, Mc Master C, Leyton L, Quest AFG. Phosphorylation of caveolin-1 on tyrosine-14 induced by ROS enhances palmitate-induced death of beta-pancreatic cells. Biochim Biophys Acta Mol Basis Dis 2015; 1852:693-708. [PMID: 25572853 DOI: 10.1016/j.bbadis.2014.12.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 12/24/2014] [Accepted: 12/27/2014] [Indexed: 01/22/2023]
Abstract
A considerable body of evidence exists implicating high levels of free saturated fatty acids in beta pancreatic cell death, although the molecular mechanisms and the signaling pathways involved have not been clearly defined. The membrane protein caveolin-1 has long been implicated in cell death, either by sensitizing to or directly inducing apoptosis and it is normally expressed in beta cells. Here, we tested whether the presence of caveolin-1 modulates free fatty acid-induced beta cell death by reexpressing this protein in MIN6 murine beta cells lacking caveolin-1. Incubation of MIN6 with palmitate, but not oleate, induced apoptotic cell death that was enhanced by the presence of caveolin-1. Moreover, palmitate induced de novo ceramide synthesis, loss of mitochondrial transmembrane potential and reactive oxygen species (ROS) formation in MIN6 cells. ROS generation promoted caveolin-1 phosphorylation on tyrosine-14 that was abrogated by the anti-oxidant N-acetylcysteine or the incubation with the Src-family kinase inhibitor, PP2 (4-amino-5-(4-chlorophenyl)-7(dimethylethyl)pyrazolo[3,4-d]pyrimidine). The expression of a non-phosphorylatable caveolin-1 tyrosine-14 to phenylalanine mutant failed to enhance palmitate-induced apoptosis while for MIN6 cells expressing the phospho-mimetic tyrosine-14 to glutamic acid mutant caveolin-1 palmitate sensitivity was comparable to that observed for MIN6 cells expressing wild type caveolin-1. Thus, caveolin-1 expression promotes palmitate-induced ROS-dependent apoptosis in MIN6 cells in a manner requiring Src family kinase mediated tyrosine-14 phosphorylation.
Collapse
Affiliation(s)
- Sergio Wehinger
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile; Research Program of Interdisciplinary Excellence in Healthy Aging (PIEI-ES), Faculty of Health Sciences, Department of Clinical Biochemistry and Immunohematology, Universidad de Talca, 3465548 Talca, Chile
| | - Rina Ortiz
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - María Inés Díaz
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Adam Aguirre
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Manuel Valenzuela
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Christopher Mc Master
- Departament of Pediatrics, Atlantic Research Centre, Dalhousie University, Halifax, NS, Canada; Department of Biochemistry and Molecular Biology, Atlantic Research Centre, Dalhousie University, Halifax, NS, Canada
| | - Lisette Leyton
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Andrew F G Quest
- Laboratory of Cellular Communication, Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile.
| |
Collapse
|
15
|
Tabassum S, Asim A, Khan RA, Arjmand F, Rajakumar D, Balaji P, Akbarsha MA. A multifunctional molecular entity Cu II–Sn IV heterobimetallic complex as a potential cancer chemotherapeutic agent: DNA binding/cleavage, SOD mimetic, topoisomerase Iα inhibitory and in vitro cytotoxic activities. RSC Adv 2015; 5:47439-47450. [DOI: 10.1039/c5ra07333b] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
New chiral l-valine-derived Schiff base complexes with the bioactive heterocyclic ligand scaffold pyrazole (Hpz) were designed and synthesized with a view to find their potential as anticancer chemotherapeutic drug candidates.
Collapse
Affiliation(s)
- Sartaj Tabassum
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Ahmad Asim
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Rais Ahmad Khan
- Department of Chemistry
- King Saud University
- Riyadh
- Kingdom of Saudi Arabia
| | - Farukh Arjmand
- Department of Chemistry
- Aligarh Muslim University
- Aligarh-202002
- India
| | - Dhivya Rajakumar
- Department of Biomedical Science
- Bharathidasan University
- Tiruchirappalli 620 024
- India
| | - Perumalsamy Balaji
- Department of Biomedical Science
- Bharathidasan University
- Tiruchirappalli 620 024
- India
| | - Mohammad Abdulkader Akbarsha
- Mahatma Gandhi-Doerenkamp Center (MGDC) for Alternatives to Use of Animals in Life Science Education
- Bharathidasan University
- Tiruchirappalli 620 024
- India
- Department of Food Science and Nutrition
| |
Collapse
|
16
|
Zainal Ariffin SH, Yeen WW, Zainol Abidin IZ, Megat Abdul Wahab R, Zainal Ariffin Z, Senafi S. Cytotoxicity effect of degraded and undegraded kappa and iota carrageenan in human intestine and liver cell lines. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:508. [PMID: 25519220 PMCID: PMC4320596 DOI: 10.1186/1472-6882-14-508] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/10/2014] [Indexed: 01/07/2023]
Abstract
Background Carrageenan is a linear sulphated polysaccharide extracted from red seaweed of the Rhodophyceae family. It has broad spectrum of applications in biomedical and biopharmaceutical field. In this study, we determined the cytotoxicity of degraded and undegraded carrageenan in human intestine (Caco-2; cancer and FHs 74 Int; normal) and liver (HepG2; cancer and Fa2N-4; normal) cell lines. Methods Food grade k-carrageenan (FGKC), dried sheet k-carrageenan (DKC), commercial grade k-carrageenan (CGKC), food grade i-carrageenan (FGIC) and commercial grade i-carrageenan (CGIC) were dissolved in hydrochloric acid and water to prepare degraded and undegraded carrageenan, respectively. Carrageenan at the concentration range of 62.5 – 2000.0 μg mL−1 was used in the study. MTT assay was used to determine the cell viability while the mode of cell death was determined by May-Grunwald Giemsa (MGG) staining, acridine orange-ethidium bromide (AO/EtBr) staining, agarose gel electrophoresis and gene expression analysis. Results Degraded FGKC, DKC and CGKC showed IC50 in 24, 48 and 72 hours treated Caco-2, FHs 74 Int, HepG2 and Fa2N-4 cell lines as tested by MTT assay. Degraded FGIC and CGIC only showed its toxicity in Fa2N-4 cells. The characteristics of apoptosis were demonstrated in degraded k-carrageenan treated Caco-2, FHs 74 Int, HepG2 and Fa2N-4 cells after MGG staining. When Caco-2 and HepG2 cells were undergone AO/EtBr staining, chromatin condensation and nuclear fragmentation were clearly seen under the microscope. However, DNA ladder was only found in HepG2 cells after gel electrophoresis analysis. Degraded k-carrageenan also inactivated PCNA, Ki-67 and survivin gene in HepG2. On the other hand, undegraded FGKC, DKC, CGKC, FGIC and CGIC treated cells showed no cytotoxic effect after analyzed by the same analyses as in degraded carrageenan. Conclusion Degraded k-carrageenan inhibited cell proliferation in Caco-2, FHs 74 Int, HepG2 and Fa2N-4 cell lines and the anti-proliferative effect was related to apoptosis together with inactivation of cell proliferating genes as determined by morphological observation and molecular analysis. However, no cytotoxic effect was found in undegraded carrageenan towards normal and cancer intestine and liver cell lines.
Collapse
|
17
|
Wu HY, Goble K, Mecha M, Wang CC, Huang CH, Guaza C, Jan TR. Cannabidiol-induced apoptosis in murine microglial cells through lipid raft. Glia 2012; 60:1182-90. [DOI: 10.1002/glia.22345] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 03/30/2012] [Indexed: 01/22/2023]
|
18
|
Rastogi P. Emergence of cancer stem cells in head and neck squamous cell carcinoma: A therapeutic insight with literature review. Dent Res J (Isfahan) 2012; 9:239-44. [PMID: 23087725 PMCID: PMC3469886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Accumulating evidence suggests that self-renewal and differentiation capabilities reside only in a subpopulation of tumor cells, termed cancer stem cells (CSCs), whereas the remaining tumor cell population lacks the ability to initiate tumor development or support continued tumor growth. In head and neck squamous cell carcinoma (HNSCC), as with other malignancies, CSCs have been increasingly shown to have an integral role in tumor initiation, disease progression, metastasis, and treatment resistance. In this article, the author summarizes the current knowledge of the role of CSCs in HNSCC and discusses the therapeutic implications and future directions of this field.
Collapse
Affiliation(s)
- Priyanka Rastogi
- Department of Oral and Maxillofacial Pathology and Microbiology, I.T.S. Center for Dental Studies and Research, Muradnagar, Ghaziabad, Uttar Pradesh, India,Address for correspondence: Dr. Priyanka Rastogi, Senior Lecturer, Department of Oral and Maxillofacial Pathology and Microbiology, I.T.S. Center for Dental Studies and Research, Muradnagar, Ghaziabad, Uttar Pradesh - 201206, India. E-mail:
| |
Collapse
|
19
|
Zhang M, Lee SJ, An C, Xu JF, Joshi B, Nabi IR, Choi AM, Jin Y. Caveolin-1 mediates Fas-BID signaling in hyperoxia-induced apoptosis. Free Radic Biol Med 2011; 50:1252-62. [PMID: 21382479 PMCID: PMC4134776 DOI: 10.1016/j.freeradbiomed.2011.02.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 02/23/2011] [Accepted: 02/25/2011] [Indexed: 12/15/2022]
Abstract
Fas-mediated apoptosis is a crucial cellular event. Fas, the Fas-associated death domain, and caspase 8 form the death-inducing signaling complex (DISC). Activated caspase 8 mediates the extrinsic pathways and cleaves cytosolic BID. Truncated BID (tBID) translocates to the mitochondria, facilitates the release of cytochrome c, and activates the intrinsic pathways. However, the mechanism causing these DISC components to aggregate and form the complex remains unclear. We found that Cav-1 regulated Fas signaling and mediated the communication between extrinsic and intrinsic pathways. Shortly after hyperoxia (4 h), the colocalization and interaction of Cav-1 and Fas increased, followed by Fas multimer and DISC formation. Deletion of Cav-1 (Cav-1-/-) disrupted DISC formation. Further, Cav-1 interacted with BID. Mutation of Cav-1 Y14 tyrosine to phenylalanine (Y14F) disrupted the hyperoxia-induced interaction between BID and Cav-1 and subsequently yielded a decreased level of tBID and resistance to hyperoxia-induced apoptosis. The reactive oxygen species (ROS) scavenger N-acetylcysteine decreased the Cav-1-Fas interaction. Deletion of glutathione peroxidase-2 using siRNA aggravated the BID-Cav-1 interaction and tBID formation. Taken together, these results indicate that Cav-1 regulates hyperoxia/ROS-induced apoptosis through interactions with Fas and BID, probably via Fas palmitoylation and Cav-1 Y14 phosphorylation, respectively.
Collapse
Affiliation(s)
- Meng Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Seon-Jin Lee
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - ChangHyeok An
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jin-fu Xu
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ivan R. Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Corresponding author. (Y. Jin)
| |
Collapse
|
20
|
Sukumari-Ramesh S, Singh N, Dhandapani KM, Vender JR. mTOR inhibition reduces cellular proliferation and sensitizes pituitary adenoma cells to ionizing radiation. Surg Neurol Int 2011; 2:22. [PMID: 21427787 PMCID: PMC3050059 DOI: 10.4103/2152-7806.77029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 01/12/2011] [Indexed: 11/10/2022] Open
Abstract
Background: Pituitary adenomas are the most frequent brain tumor in adults. Although histologically benign, pituitary tumors cause significant morbidity and mortality. Neurosurgery and medical therapeutics may lessen the morbidity and mortality associated with pituitary tumors; however, these treatments are associated with significant adverse side effects. Thus, an improved understanding of pituitary adenomas at the molecular and cellular level is needed to design novel therapeutic compounds. Methods: To assess the effect of mammalian target of rapamycin (mTOR) inhibition on pituitary adenoma cells, rat GH3 or MMQ cells were treated with the clinically useful mTOR inhibitors, rapamycin or RAD001. Cellular proliferation and growth following exposure to mTOR inhibitors or radiation were assessed using biochemical methods. Results: In the present study, we observed basal activation of mTOR, downstream of constitutive Akt signaling, in rat GH3 adenoma cells. Functionally, the mTOR inhibitors, rapamycin and RAD001 (500 pM–5 nM), induced G1 growth arrest within 24 hours, an effect associated with reduced cellular proliferation. Both rapamycin and RAD001 decreased the phosphorylation of mTOR at the serine 2448, a key determinant of mTOR activity. Inhibition of mTOR also radiosensitized GH3 cells such that 2.5 Gy in combination with 500 pM rapamycin or RAD001 reduced cellular viability more effectively than 2.5 or 10 Gy alone. Conclusions: These data may support a possible therapeutic role for mTOR inhibitors in limiting the cellular proliferation and radioresistance of pituitary adenoma cells.
Collapse
|
21
|
Huang MH, Shen AY, Wang TS, Wu HM, Kang YF, Chen CT, Hsu TI, Chen BS, Wu SN. Inhibitory action of methadone and its metabolites on erg-mediated K+ current in GH₃ pituitary tumor cells. Toxicology 2010; 280:1-9. [PMID: 21094671 DOI: 10.1016/j.tox.2010.10.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 10/20/2010] [Accepted: 10/21/2010] [Indexed: 01/05/2023]
Abstract
Methadone (Mtd) is a widely used opioid drug associated with the side effect of hyperprolactinemia. The mechanism of how Mtd induces prolactin secretion remains unclear. The effects of Mtd and its two main metabolites (EDDP: (±)-2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolinium percholarate and EMDP: 2-ethyl-5-methyl-3,3-dipnehyl-1-pyrroline) on ion currents were investigated in GH₃ pituitary tumor cells. Hyperpolarization-elicited K+ currents in GH₃ cells bathed in a high-K(+), Ca(2+)-free solution were studied to evaluate the effects of Mtd and other related compounds on the ether-à-go-go-related-gene (erg) K(+) current (I(K(erg))). Mtd suppressed the amplitude of I(K(erg)) in a concentration-dependent manner with an IC(50) value of 10.4 μM. With the aid of a minimal binding scheme, the inhibitory action of Mtd on I(K(erg)) was estimated with a dissociation constant of 8.2 μM. Mtd tended to increase the rate of I(K(erg)) deactivation in a voltage-dependent fashion. EDDP (10 μM) had no effect on I(K(erg)), while EMDP (10μM) slightly suppressed it. In GH₃ cells incubated with naloxone (30 μM), the Mtd-induced inhibition of I(K(erg)) remained unaltered. Under cell-attached voltage-clamp recordings, Mtd increased the frequency of spontaneous action currents with no change in current amplitude. Similarly, Mtd can suppress I(K(erg)) in differentiated NG108-15 cells; dynorphin A(1-13) did not reverse Mtd-induced inhibition of I(K(erg)). This study shows that Mtd has a depressant effect on I(K(erg)), and suggests its ability to affect membrane excitability and prolactin secretion. The cyclization of Mtd, in which EDDP and EMDP are formed, tends to be critical in removal of the Mtd binding to erg K+ channel.
Collapse
Affiliation(s)
- Mei-Han Huang
- College of Medical and Health Sciences, Fooyin University, Ta-Liao, Kaohsiung County, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ifere GO, Equan A, Gordon K, Nagappan P, Igietseme JU, Ananaba GA. Cholesterol and phytosterols differentially regulate the expression of caveolin 1 and a downstream prostate cell growth-suppressor gene. Cancer Epidemiol 2010; 34:461-71. [PMID: 20466611 DOI: 10.1016/j.canep.2010.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Revised: 04/10/2010] [Accepted: 04/12/2010] [Indexed: 01/18/2023]
Abstract
BACKGROUND The purpose of our study was to show the distinction between the apoptotic and anti-proliferative signaling of phytosterols and cholesterol-enrichment in prostate cancer cell lines, mediated by the differential transcription of caveolin-1, and N-myc downstream-regulated gene 1 (NDRG1), a pro-apoptotic androgen-regulated tumor suppressor. METHODS PC-3 and DU145 cells were treated with sterols (cholesterol and phytosterols) for 72h, followed by trypan blue dye-exclusion measurement of necrosis and cell growth measured with a Coulter counter. Sterol induction of cell growth-suppressor gene expression was evaluated by mRNA transcription using RT-PCR, while cell cycle analysis was performed by FACS analysis. Altered expression of Ndrg1 protein was confirmed by Western blot analysis. Apoptosis was evaluated by real time RT-PCR amplification of P53, Bcl-2 gene and its related pro- and anti-apoptotic family members. RESULTS Physiological doses (16microM) of cholesterol and phytosterols were not cytotoxic in these cells. Cholesterol-enrichment promoted cell growth (P<0.05), while phytosterols significantly induced growth-suppression (P<0.05) and apoptosis. Cell cycle analysis showed that contrary to cholesterol, phytosterols decreased mitotic subpopulations. We demonstrated for the first time that cholesterols concertedly attenuated the expression of caveolin-1 (cav-1) and NDRG1 genes in both prostate cancer cell lines. Phytosterols had the opposite effect by inducing overexpression of cav-1, a known mediator of androgen-dependent signals that presumably control cell growth or apoptosis. CONCLUSIONS Cholesterol and phytosterol treatment differentially regulated the growth of prostate cancer cells and the expression of p53 and cav-1, a gene that regulates androgen-regulated signals. These sterols also differentially regulated cell cycle arrest, downstream pro-apoptotic androgen-regulated tumor suppressor, NDRG1 suggesting that cav-1 may mediate pro-apoptotic NDRG1 signals. Elucidation of the mechanism for sterol modulation of growth and apoptosis signaling may reveal potential targets for cancer prevention and/or chemotherapeutic intervention. Sterol regulation of NDRG1 transcription suggests its potential as biomarker for prediction of neoplasms that would be responsive to chemoprevention by phytosterols.
Collapse
Affiliation(s)
- Godwin O Ifere
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Vender JR, Laird MD, Dhandapani KM. Inhibition of NFkappaB reduces cellular viability in GH3 pituitary adenoma cells. Neurosurgery 2008; 62:1122-7; discussion 1027-8. [PMID: 18580810 DOI: 10.1227/01.neu.0000325874.82999.75] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Adenomas of the pituitary gland are among the most common types of tumors of the adult brain. Although adenomas are histologically benign, they may be associated with significant morbidity and mortality, mostly because of their invasive growth pattern and hormone hypersecretion. Current medical therapies are suppressive, acting at a receptor level. Thus, there is a need to identify novel cellular and molecular targets for pituitary tumors. We investigated the possible role of the NFkappaB transcription factor in pituitary tumor cell growth. METHODS The effect of NFkappaB pathway inhibition on cellular viability was studied in the GH3 pituitary adenoma cell line, a well-characterized rat cell line that secretes growth hormone and prolactin. Cells were treated with mechanistically diverse pharmacological NFkappaB pathway inhibitors or with molecular inhibitors that were overexpressed in tumor cells before the assessment of cellular viability. NFkappaB activity was also assessed in GH3 cells using deoxyribonucleic acid binding assays. RESULTS GH3 cells exhibited constitutive NFkappaB activity, which contributed to increased cellular proliferation. Treatment with wedelolactone, an IkappaB kinase inhibitor, or overexpression of an IkappaB super-repressor reduced cell viability, further implicating NFkappaB in pituitary tumor cell growth. Pharmacological or molecular inhibition of Akt similarly reduced GH3 viability and NFkappaB binding, suggesting that constitutive activation of NFkappaB may be, at least in part, mediated by Akt. CONCLUSION Directed targeting of the Akt and NFkappaB signaling pathways may be a useful adjunct in the clinical management of pituitary tumors. Further elucidation of this pathway may yield novel information regarding the behavior of pituitary tumors in humans.
Collapse
Affiliation(s)
- John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | |
Collapse
|
24
|
Vender JR, Laird MD, Dhandapani KM. INHIBITION OF NFκB REDUCES CELLULAR VIABILITY IN GH3 PITUITARY ADENOMA CELLS. Neurosurgery 2008. [DOI: 10.1227/01.neu.0000312715.01310.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
25
|
Xia T, Kovochich M, Liong M, Zink JI, Nel AE. Cationic polystyrene nanosphere toxicity depends on cell-specific endocytic and mitochondrial injury pathways. ACS NANO 2008; 2:85-96. [PMID: 19206551 DOI: 10.1021/nn700256c] [Citation(s) in RCA: 476] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The exponential increase in the number of new nanomaterials that are being produced increases the likelihood of adverse biological effects in humans and the environment. In this study we compared the effects of cationic nanoparticles in five different cell lines that represent portal-of-entry or systemic cellular targets for engineered nanoparticles. Although 60 nm NH(2)-labeled polystyrene (PS) nanospheres were highly toxic in macrophage (RAW 264.7) and epithelial (BEAS-2B) cells, human microvascular endothelial (HMEC), hepatoma (HEPA-1), and pheochromocytoma (PC-12) cells were relatively resistant to particle injury. While the death pathway in RAW 264.7 cells involves caspase activation, the cytotoxic response in BEAS-2B cells is more necrotic in nature. Using fluorescent-labeled NH(2)-PS, we followed the routes of particle uptake. Confocal microscopy showed that the cationic particles entered a LAMP-1 positive lysosomal compartment in RAW 264.7 cells from where the particles could escape by lysosomal rupture. A proton pump inhibitor interfered in this pathway. Subsequent deposition of the particles in the cytosol induced an increase in mitochondrial Ca(2+) uptake and cell death that could be suppressed by cyclosporin A (CsA). In contrast, NH(2)-PS toxicity in BEAS-2B cells did not involve the LAMP-1 endosomal compartment, stimulation of proton pump activity, or an increase in mitochondrial Ca(2+). Particles were taken up by caveolae, and their toxicity could be disrupted by cholesterol extraction from the surface membrane. Although the particles induced mitochondrial damage and ATP depletion, CsA did not affect cytotoxicity. Cationic particles were taken up into HEPA-1, HMEC, and PC-12 cells, but this did not lead to lysosomal permeabilization, increased Ca(2+) flux, or mitochondrial damage. Taken together, the results of this study demonstrate the importance of cell-specific uptake mechanisms and pathways that could lead to sensitivity or resistance to cationic particle toxicity.
Collapse
Affiliation(s)
- Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|