301
|
Léger H, Santana E, Leu NA, Smith ET, Beltran WA, Aguirre GD, Luca FC. Ndr kinases regulate retinal interneuron proliferation and homeostasis. Sci Rep 2018; 8:12544. [PMID: 30135513 PMCID: PMC6105603 DOI: 10.1038/s41598-018-30492-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 08/01/2018] [Indexed: 12/31/2022] Open
Abstract
Ndr2/Stk38l encodes a protein kinase associated with the Hippo tumor suppressor pathway and is mutated in a naturally-occurring canine early retinal degeneration (erd). To elucidate the retinal functions of Ndr2 and its paralog Ndr1/Stk38, we generated Ndr1 and Ndr2 single knockout mice. Although retinal lamination appeared normal in these mice, Ndr deletion caused a subset of Pax6-positive amacrine cells to proliferate in differentiated retinas, while concurrently decreasing the number of GABAergic, HuD and Pax6-positive amacrine cells. Retinal transcriptome analyses revealed that Ndr2 deletion increased expression of neuronal stress genes and decreased expression of synaptic organization genes. Consistent with the latter, Ndr deletion dramatically reduced levels of Aak1, an Ndr substrate that regulates vesicle trafficking. Our findings indicate that Ndr kinases are important regulators of amacrine and photoreceptor cells and suggest that Ndr kinases inhibit the proliferation of a subset of terminally differentiated cells and modulate interneuron synapse function via Aak1.
Collapse
Affiliation(s)
- Hélène Léger
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Evelyn Santana
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - N Adrian Leu
- Center for Animal Transgenesis and Germ Cell Research, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Eliot T Smith
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - William A Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States
| | - Francis C Luca
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, United States.
| |
Collapse
|
302
|
Respondek M, Beberok A, Rok J, Rzepka Z, Wrześniok D, Buszman E. MIM1, the Mcl-1 - specific BH3 mimetic induces apoptosis in human U87MG glioblastoma cells. Toxicol In Vitro 2018; 53:126-135. [PMID: 30134184 DOI: 10.1016/j.tiv.2018.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/04/2018] [Accepted: 08/16/2018] [Indexed: 01/19/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and high aggressive malignant brain tumor. Despite evolving oncology treatment and novel chemotherapeutic agents the median survival of patients diagnosed with GBM is only 12-15 months. This grim fact highlights necessity to identify new drugs that could improve the effectiveness of GBM patients treatment. MIM1 is a specific low molecular Mcl-1 protein inhibitor able to induce Mcl-1-dependent cancer cells death. The aim of this study was to examine the effect of MIM1 as well as MIM1 and temozolomide (TMZ) mixture on cell viability, apoptosis and cell cycle progression in human U87MG glioblastoma cells. Cell viability was performed by the WST-1 assay. Mitochondrial membrane potential, Annexin V assay, DNA fragmentation and cell cycle distribution were determined by fluorescence image cytometer NucleoCounter NC-3000. The obtained results show that MIM1 and MIM1/TMZ mixture decrease glioblastoma cells viability in a dose- and time- dependent manner. Moreover, the exposure of U87MG cells to MIM1 and MIM1/TMZ mixture causes mitochondrial dysfunction as well as DNA fragmentation and cell cycle arrest at G2/M phase. This study provides for the first time convincing evidence that BH3 mimetic MIM1, which inhibits Mcl-1 antiapoptotic protein may be an efficacious molecule able to induction of apoptosis and sensitize GBM cells to alkylating agents.
Collapse
Affiliation(s)
- Michalina Respondek
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland.
| | - Artur Beberok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Jakub Rok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Zuzanna Rzepka
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Ewa Buszman
- Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Department of Pharmaceutical Chemistry, Jagiellońska 4, 41-200 Sosnowiec, Poland
| |
Collapse
|
303
|
Sitarek P, Synowiec E, Kowalczyk T, Śliwiński T, Skała E. An In Vitro Estimation of the Cytotoxicity and Genotoxicity of Root Extract from Leonurus sibiricus L. Overexpressing AtPAP1 against Different Cancer Cell Lines. Molecules 2018; 23:molecules23082049. [PMID: 30115821 PMCID: PMC6222913 DOI: 10.3390/molecules23082049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/08/2018] [Accepted: 08/12/2018] [Indexed: 12/22/2022] Open
Abstract
As the current cancer treatment success rate is not sufficient, interest has grown in plants as possible sources of anti-cancer compounds. One such plant with a broad spectrum of activity is Lenourus sibiricus of the family Lamiaceae. This study investigates for the first time both the genotoxic and cytotoxic activities of TR (transformed) and AtPAP1 TR (with over-expression of transcriptional factor) root extracts of Lenourus sibiricus against various cancer cell lines (CCRF-CEM, K-562 and A549). Both tested extracts showed a cytotoxic effect on CCRF-CEM and K-562 cell lines, but strongest activity was observed for the AtPAP1 TR extract. No cytotoxic effect was observed against the A549 cell line in the tested concentration range, and it was found that both tested extracts may induce apoptosis by decreasing mitochondrial membrane potential and inducing nDNA damage lesion in the TP53 region and mtDNA in ND1 (mitochondrially encoded NADH: ubiquinone oxidoreductase core subunit 1) and ND5 (mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 5) regions in K-562 and CCRF-CEM. Our results confirmed that TR and AtPAP1 TR root extracts from L. sibiricus are cytotoxic and genotoxic against different model cell lines (CCRF-CEM and K-562). However, the observed genotoxicity of both extracts needs to be confirmed by additional studies. These preclinical observations support the use of L. sibiricus with other pharmacological purposes.
Collapse
Affiliation(s)
- Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland.
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| | - Tomasz Kowalczyk
- Department of Genetics and Plant Molecular Biology and Biotechnology, The University of Łódź, Banacha 12/13, 90-237 Łódź, Poland.
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska 141/143, 90-236 Łódź, Poland.
| | - Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland.
| |
Collapse
|
304
|
Bauer E, Domingo X, Balcells C, Polat IH, Crespo M, Quirante J, Badía J, Baldomà L, Font-Bardia M, Cascante M. Synthesis, characterization and biological activity of new cyclometallated platinum(iv) iodido complexes. Dalton Trans 2018; 46:14973-14987. [PMID: 29048088 DOI: 10.1039/c7dt03448b] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The synthesis of six novel cyclometallated platinum(iv) iodido complexes is accomplished by intermolecular oxidative addition of methyl iodide (compounds 2a-2c) or iodine (compounds 3a-3c) upon cyclometallated platinum(ii) compounds [PtX{(CH3)2N(CH2)3NCH(4-ClC6H3)}] (1a-1c: X = Cl, CH3 or I). The X-ray molecular structures of platinum(ii) compound 1c and platinum(iv) compounds 3b and 3a' (an isomer of 3a) are reported. The cytotoxic activity against a panel of human adenocarcinoma cell lines (A-549 lung, MDA-MB-231 and MCF-7 breast, and HCT-116 colon), DNA interaction, topoisomerase I, IIα, and cathepsin B inhibition, and cell cycle arrest, apoptosis and ROS generation of the investigated complexes are presented. Remarkable antiproliferative activity was observed for most of the synthesized cycloplatinated compounds (series 1-3) in all the selected carcinoma cell lines. The best inhibition was provided for the octahedral platinum(iv) compounds 2a-2c exhibiting a methyl and an iodido axial ligand. Preliminary biological results point to a different mechanism of action for the investigated compounds. Cyclometallated platinum(ii) compounds 1a-1c modify the DNA migration as cisplatin. In contrast, cyclometallated platinum(iv) compounds 2a-2c and 3a-3c did not modify the DNA tertiary structure neither in the absence nor in the presence of ascorbic acid, which made them incapable of reducing platinum(iv) compounds 2b and 2c in a buffered aqueous medium (pH 7.40) according to 1H NMR experiments. Remarkable topoisomerase IIα inhibitory activity is reported for platinum(iv) complexes 2b and 3a and in addition, for the last one, a moderate cathepsin B inhibition is reported. Cell cycle arrest (decrease in G0/G1 and G2 phases and arrest in the S phase), induction of apoptosis and ROS generation are related to the antiproliferative activity of some representative octahedral cyclometallated platinum(iv) compounds (2b and 2c).
Collapse
Affiliation(s)
- Emma Bauer
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Facultat de Química, Universitat de Barcelona, Diagonal 645, 08028-Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
305
|
Cunningham KE, Novak EA, Vincent G, Siow VS, Griffith BD, Ranganathan S, Rosengart MR, Piganelli JD, Mollen KP. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation contributes to the pathogenesis of experimental colitis via inhibition of intestinal epithelial cell proliferation. FASEB J 2018; 33:1330-1346. [PMID: 30113881 DOI: 10.1096/fj.201800535r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The incidence and prevalence of inflammatory bowel disease (IBD) are increasing worldwide. IBD is known to be multifactorial, but inflammatory signaling within the intestinal epithelium and a subsequent failure of the intestinal epithelial barrier have been shown to play essential roles in disease pathogenesis. CaMKIV is a multifunctional protein kinase associated with inflammation and cell cycle regulation. CaMKIV has been extensively studied in autoimmune diseases, but a role in idiopathic intestinal inflammation has not been described. In this study, active CaMKIV was highly expressed within the intestinal epithelium of humans with ulcerative colitis and wild-type (WT) mice with experimental induced colitis. Clinical disease severity directly correlates with CaMKIV activation, as does expression of proinflammatory cytokines and histologic features of colitis. In WT mice, CaMKIV activation is associated with increases in expression of 2 cell cycle proarrest signals: p53 and p21. Cell cycle arrest inhibits proliferation of the intestinal epithelium and ultimately results in compromised intestinal epithelial barrier integrity, further perpetuating intestinal inflammation during experimental colitis. Using a CaMKIV null mutant mouse, we demonstrate that a loss of CaMKIV protects against murine DSS colitis. Small molecules targeting CaMKIV activation may provide therapeutic benefit for patients with IBD.-Cunningham, K. E., Novak, E. A., Vincent, G., Siow, V. S., Griffith, B. D., Ranganathan, S., Rosengart, M. R., Piganelli, J. D., Mollen, K. P. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation contributes to the pathogenesis of experimental colitis via inhibition of intestinal epithelial cell proliferation.
Collapse
Affiliation(s)
- Kellie E Cunningham
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elizabeth A Novak
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Garret Vincent
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Vei Shaun Siow
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Brian D Griffith
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarangarajan Ranganathan
- Department of Pathology, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jon D Piganelli
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin P Mollen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
| |
Collapse
|
306
|
Yurdacan B, Egeli U, Eskiler GG, Eryilmaz IE, Cecener G, Tunca B. The role of usnic acid-induced apoptosis and autophagy in hepatocellular carcinoma. Hum Exp Toxicol 2018; 38:201-215. [DOI: 10.1177/0960327118792052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Usnic acid (UA) is a multifunctional bioactive lichen secondary metabolite with potential anti-cancer properties. Although the promising therapeutic effects of UA have been investigated in different cancer cell lines, the mechanism driving UA-induced cell death has yet to be elucidated. As the type of cell death (apoptosis or autophagy) induced by UA may vary depending on the cancer cell type, we first studied the cytotoxic effects of UA in HEPG2 (HBV(−)) and SNU-449(HBV(+)) hepatocellular carcinoma (HCC) cell lines. HCC cell viability was considerably reduced in a dose-dependent manner at 12, 24, and 48 h after treatment with UA ( p < 0.05). However, SNU-449 cells were more sensitive to UA than HEPG2 cells. UA also induced apoptotic cell death in HCC cells with cell cycle arrest at G0/G1 and G2/M phase depending on the genetic profile of each cell type. On the other hand, we observed acidic vesicular organelles in HCC cells after 36 h of UA treatment. Taken together, these findings suggest that UA stimulates apoptosis and autophagy in HEPG2 and SNU-449 cells without damaging normal control cells. Thus, UA might be a potential therapeutic compound for HCC treatment. However, there is a need for further studies investigating the death-promoting or preventing roles for autophagy and the molecular signaling mechanisms induced by UA treatment.
Collapse
Affiliation(s)
- B Yurdacan
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - U Egeli
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - G Guney Eskiler
- Department of Medical Biology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - IE Eryilmaz
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - G Cecener
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - B Tunca
- Department of Medical Biology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
307
|
The combination of epigenetic drugs SAHA and HCI-2509 synergistically inhibits EWS-FLI1 and tumor growth in Ewing sarcoma. Oncotarget 2018; 9:31397-31410. [PMID: 30140378 PMCID: PMC6101143 DOI: 10.18632/oncotarget.25829] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022] Open
Abstract
Purpose Epigenetic regulation is crucial in mammalian development and maintenance of tissue-cell specific functions. Perturbation of epigenetic balance may lead to alterations in gene expression, resulting in cellular transformation and malignancy. Previous studies in Ewing sarcoma (ES) have shown that the Nucleosome Remodeling Deacetylase (NuRD) complex binds directly to EWS-FLI1 oncoprotein and modulates its transcriptional activity. The role of EWS-FLI1 as a driver of proliferation and transformation in ES is widely known, but the effect of epigenetic drugs on fusion activity remains poorly described. The present study evaluated the combination effects of the histone deacetylases inhibitor suberoylanilide hydroxamic acid (SAHA) and Lysine-specific demethylase1 inhibitor (HCI-2509) on different biological functions in ES and in comparison to monotherapy treatments. Results The study of proliferation and cell viability showed a synergistic effect in most ES cell lines analyzed. An enhanced effect was also observed in the induction of apoptosis, together with accumulation of cells in G1 phase and a blockage of the migratory capacity of ES cell lines. Treatment, either in monotherapy or in combination, caused a significant decrease of EWS-FLI1 mRNA and protein levels and this effect is mediated in part by fusion gene promoter regulation. The anti-tumor effect of this combination was confirmed in patient-derived xenograft mouse models, in which only the combination treatment led to a statistically significant decrease in tumor volume. Conclusions The combination of SAHA and HCI-2509 is proposed as a novel treatment strategy for ES patients to inhibit the essential driver of this sarcoma and tumor growth.
Collapse
|
308
|
Design and synthesis of new antitumor agents with the 1,7-epoxycyclononane framework. Study of their anticancer action mechanism by a model compound. Bioorg Med Chem 2018; 26:3379-3398. [PMID: 29784272 DOI: 10.1016/j.bmc.2018.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/13/2018] [Accepted: 05/08/2018] [Indexed: 11/23/2022]
Abstract
This article describes the design, synthesis and biological evaluation of a new family of antitumor agents having the 1,7-epoxycyclononane framework. We have developed a versatile synthetic methodology that allows the preparation of a chemical library with structural diversity and in good yield. The synthetic methodology has been scaled up to the multigram level and can be developed in an enantioselective fashion. The study in vitro of a model compound, in front of the cancer cell lines HL-60 and MCF-7, showed a growth inhibitory effect better than that of cisplatin. The observation of cancer cells by fluorescence microscopy showed the presence of apoptotic bodies and a degradation of microtubules. The study of cell cycle and mechanism of death of cancer cells by flow cytometry indicates that the cell cycle arrested at the G0/G1 phase and that the cells died by apoptosis preferably over necrosis. A high percentage of apoptotic cells at the subG0/G1 level was observed. This indicates that our model compound does not behave as an antimitotic agent like nocodazole, used as a reference, which arrests the cell cycle at G2/M phase. The interaction of anticancer agents with DNA molecules was evaluated by atomic force microscopy, circular dichroism and electrophoresis on agarose gel. The results indicate that the model compound has not DNA as a target molecule. The in silico study of the model compound showed a potential good oral bioavailability.
Collapse
|
309
|
Mesbahi Y, Zekri A, Ghaffari SH, Tabatabaie PS, Ahmadian S, Ghavamzadeh A. Blockade of JAK2/STAT3 intensifies the anti-tumor activity of arsenic trioxide in acute myeloid leukemia cells: Novel synergistic mechanism via the mediation of reactive oxygen species. Eur J Pharmacol 2018; 834:65-76. [PMID: 30012499 DOI: 10.1016/j.ejphar.2018.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/02/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are essential mediators of crucial cellular processes including apoptosis, proliferation, survival and cell cycle. Their regulatory role in cancer progression has seen in different human malignancies such as acute myeloid leukemia (AML). AML patients suffer from high resistance of the tumors against routine therapeutics including ATO. ATO enhance reactive oxygen species levels and induce apoptosis and suppresses proliferation in AML cells. However, some pathways such as JAK2/STAT3 ease anti-tumor activity of ATO by reducing reactive oxygen species amount and protecting the cell from apoptosis. In the present study, we use ruxolitinib (potent JAK2 inhibitor) to increase the sensitivity of AML cells to ATO treatment. We test, the effect of this combination on metabolic activity, proliferation, colony formation, cell cycle distribution, apoptosis, oxidative stress and DNA damage. Our results showed that combination of ATO with ruxolitinib synergistically reduced metabolic activity, proliferation and survival of AML cell lines. This combination induced G1/S cell cycle arrest because of reactive oxygen species elevation and GSH reduction. Besides, enhancement of reactive oxygen species increased apoptosis rate in combination samples. We uncovered that the synergistic anti-tumor effect of ATO and ruxolitinib in AML cells mediates via reactive oxygen species elevation and DNA damage. Overall, our results show that the combinatorial therapy of AML cells is more effective than solo-targeted therapy.
Collapse
Affiliation(s)
- Yashar Mesbahi
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran; Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Ali Zekri
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Seyed H Ghaffari
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran; Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran.
| | | | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran; Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| |
Collapse
|
310
|
Mirza-Aghazadeh-Attari M, Darband SG, Kaviani M, Mihanfar A, Aghazadeh Attari J, Yousefi B, Majidinia M. DNA damage response and repair in colorectal cancer: Defects, regulation and therapeutic implications. DNA Repair (Amst) 2018; 69:34-52. [PMID: 30055507 DOI: 10.1016/j.dnarep.2018.07.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/15/2018] [Accepted: 07/15/2018] [Indexed: 12/11/2022]
Abstract
DNA damage response, a key factor involved in maintaining genome integrity and stability, consists of several kinase-dependent signaling pathways, which sense and transduce DNA damage signal. The severity of damage appears to determine DNA damage responses, which can include cell cycle arrest, damage repair and apoptosis. A number of recent studies have demonstrated that defection in signaling through this network is thought to be an underlying mechanism behind the development and progression of various types of human malignancies, including colorectal cancer. In this review, colorectal cancer and its molecular pathology as well as DNA damage response is briefly introduced. Finally, the involvement of key components of this network in the initiation/progression, prognosis, response to treatment and development of drug resistance is comprehensively discussed.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saber Ghazizadeh Darband
- Danesh Pey Hadi Co., Health Technology Development Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | - Ainaz Mihanfar
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
311
|
Kee JY, Han YH, Mun JG, Um JY, Hong SH. Pharmacological effect of prohibited combination pair Panax ginseng and Veratrum nigrum on colorectal metastasis in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2018; 220:177-187. [PMID: 29601980 DOI: 10.1016/j.jep.2018.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/21/2018] [Accepted: 03/26/2018] [Indexed: 05/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The pharmacological effect derived from herb-herb interaction is important to constitute the prescription especially in traditional oriental medicine. The relationship of two medicinal herbs is called "couplet medicinals" which is used in pair for the purpose of enhancing the therapeutic effect, reducing the toxic effect or the adverse effect. The "Eighteen Incompatible Medicaments" constitute one of the contents in the incompatibility of traditional oriental drugs in a prescription. Among the "Eighteen Incompatible Medicaments", the roots and rhizomes of Veratrum nigrum (VN), is incompatible with the roots and rhizomes of Panax ginseng (PG). However, definite evidences of adverse effect by these combinations has yet to be reported. MATERIALS AND METHODS The aim of the present study was to investigate the effects of ethanol extracts of PG, VN, and their combination (P + V) on the metastatic ability of colorectal cancer (CRC) cells using WST assay, flow cytometry, western blot analysis, real-time RT-PCR, immunofluorescence, migration assay, invasion assay, zymography, and an in vivo experiment with a lung-metastasis mouse model. RESULTS The PG extract decreased cell proliferation by inducing cell cycle arrest and apoptosis of CRC cells. In addition, PG inhibited metastatic abilities of CRC cells including Epithelial-Mesenchymal Transition, migration, and invasion. Additionally, the PG extract suppressed lung metastasis of the CRC cells in the mouse model. However, the P + V extract exhibited weaker anti-proliferative and anti-metastatic effects than PG alone. CONCLUSION Based on these results, the P + V couplet medicinal attenuates the anti-metastatic effects of PG, both in vitro and in vivo.
Collapse
Affiliation(s)
- Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jeong-Geon Mun
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea.
| |
Collapse
|
312
|
da Silva RF, Dhar D, Raina K, Kumar D, Kant R, Cagnon VHA, Agarwal C, Agarwal R. Nintedanib inhibits growth of human prostate carcinoma cells by modulating both cell cycle and angiogenesis regulators. Sci Rep 2018; 8:9540. [PMID: 29934570 PMCID: PMC6014981 DOI: 10.1038/s41598-018-27831-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignancy and second leading cause of cancer-related deaths in American men. Proliferating cells have higher need for nutrients and oxygen, triggering angiogenesis that plays a critical role in tumor growth, progression and metastasis. Consequently, immense focus has converged onto inhibitors of angiogenesis in cancer treatment, such as Nintedanib, which has shown exceptional antitumor activity via inhibiting cell proliferation and the resulting tumor growth, primarily due to its combined action on tumor cells, endothelial cells and pericytes. Accordingly, here we assessed both in vitro and in vivo efficacy of Nintedanib in PCa. The results showed that Nintedanib decreased cell viability in both androgen dependent- and -independent PCa cells, together with a decrease in cell motility and invasiveness. Nintedanib also reduced the expression of significant genes responsible for cell cycle progression. PCa PC3 xenograft-carrying nude mice treated with Nintedanib showed significantly decreased tumor volume and cell proliferation alongside diminished levels of pro-angiogenic molecules and blood vessel densities. In conclusion, we report that Nintedanib has strong efficacy against PCa in pre-clinical models via modulation of various pathways, and that it could be employed as a promising new strategy to manage PCa clinically.
Collapse
Affiliation(s)
- Raquel Frenedoso da Silva
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Deepanshi Dhar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dileep Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Valeria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA. .,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
313
|
Noor A, Umelo IA, Kronenberger P, Giron P, De Vlieghere E, De Wever O, Teugels E, De Grève J. Targeting Polo-like kinase 1 and TRAIL enhances apoptosis in non-small cell lung cancer. Oncotarget 2018; 9:28731-28744. [PMID: 29983892 PMCID: PMC6033352 DOI: 10.18632/oncotarget.25618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/31/2018] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis in cancer cells without causing damage to normal cells. However, some tumors are resistant to TRAIL monotherapy and clinical studies assessing targeted agents towards the TRAIL receptor have failed to show robust therapeutic activity. Evidence has shown that standard anti-mitotic drugs can induce synergistic apoptosis upon combination with TRAIL via cell cycle arrest. Polo like kinase-1 (PLK1) plays a critical role in different stages of cell cycle progression and mitosis. A number of investigations have demonstrated that PLK1 inhibition causes cell cycle arrest and mitotic catastrophe in non-small cell lung cancer (NSCLC), and we thus postulated that PLK1 inhibition could enhance TRAIL-induced apoptosis. We demonstrate that the combination of a TRAIL receptor agonist and a PLK1 inhibitor synergistically reduces cell viability, and strongly increases apoptosis in NSCLC cellular models. Consistent with our in vitro observations, this drug combination also significantly reduces tumor growth in vivo. Our data additionally reveal that G2/M cell cycle arrest and downregulation of Mcl-1 and signal transducer and activator of transcription 3 (STAT3) activity following PLK1 inhibition may contribute to the sensitization of TRAIL-induced apoptosis in NSCLC. Together, these data support the further exploration of combined TRAIL and PLK1 inhibition in the treatment of NSCLC.
Collapse
Affiliation(s)
- Alfiah Noor
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ijeoma Adaku Umelo
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Peter Kronenberger
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Laboratory of Biotechnology, Department of Healthcare, Erasmushogeschool Brussel, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Erik Teugels
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jacques De Grève
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
314
|
Sakthivel R, Malar DS, Devi KP. Phytol shows anti-angiogenic activity and induces apoptosis in A549 cells by depolarizing the mitochondrial membrane potential. Biomed Pharmacother 2018; 105:742-752. [PMID: 29908495 DOI: 10.1016/j.biopha.2018.06.035] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/25/2022] Open
Abstract
In the present study, the antiproliferative activity of phytol and its mechanism of action against human lung adenocarcinoma cell line A549 were studied in detail. Results showed that phytol exhibited potent antiproliferative activity against A549 cells in a dose and time-dependent manner with an IC50 value of 70.81 ± 0.32 μM and 60.7 ± 0.47 μM at 24 and 48 h, respectively. Phytol showed no adverse toxic effect in normal human lung cells (L-132), but mild toxic effect was observed when treated with maximum dose (67 and 84 μM). No membrane-damaging effect was evidenced by PI staining and SEM analysis. The results of mitochondrial membrane potential analysis, cell cycle analysis, FT-IR and Western blotting analysis clearly demonstrated the molecular mechanism of phytol as induction of apoptosis in A549 cells, as evidenced by formation of shrinked cell morphology with membrane blebbing, depolarization of mitochondrial membrane potential, increased cell population in the sub-G0 phase, band variation in the DNA and lipid region, downregulation of Bcl-2, upregulation of Bax and the activation of caspase-9 and -3. In addition, phytol inhibited the CAM vascular growth as evidenced by CAM assay, which positively suggests that phytol has anti-angiogenic potential. Taken together, these findings clearly demonstrate the mode of action by which phytol induces cell death in A549 lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Ravi Sakthivel
- Department of Biotechnology, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Dicson Sheeja Malar
- Department of Biotechnology, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi 630 003, Tamil Nadu, India.
| |
Collapse
|
315
|
Prasad N, Sabarwal A, Yadav UCS, Singh RP. Lupeol induces S-phase arrest and mitochondria-mediated apoptosis in cervical cancer cells. J Biosci 2018; 43:249-261. [PMID: 29872014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cervical cancer is fourth most common fatal cancer in women worldwide. Lupeol is a dietary triterpenoid and has shown its anticancer efficacy against various cancer types with selectivity in targeting cancer cells. In the present study, anticancer efficacy and mechanism of action of a phytochemical, lupeol, in human cervical carcinoma (HeLa) cells has been examined. The anticancer efficacy of lupeol was assessed by trypan blue cell counting, annexin Vassay, cell cycle analysis, expression of apoptotic proteins by RT-PCR and Western blotting and assessment of mitochondrial ROS generation by mitosox and mitotracker assays. Our results demonstrated that lupeol decreased cell proliferation and viability of HeLa cells significantly (p less than 0.001). Lupeol induced S-phase cell cycle arrest and also decreased the expression of S-phase Cyclins and CDKs and increased the expression of cyclin-dependent kinase inhibitors, p21 at transcriptional and translational level. Further, lupeol induced apoptosis and increased the expression of apoptosis markers such as cleaved PARP and Bax:Bcl-2 ratio. Furthermore, mitosox and mitotracker dye incubation followed by FACS analysis showed an increase in mitochondrial superoxide generation and reduction in healthy mitochondrial mass. These results suggest that lupeol could be an effective chemotherapeutic agent against cervical carcinoma due to its growth inhibitory activity through induction of S-phase cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Nupoor Prasad
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | | | | | | |
Collapse
|
316
|
Cheah FK, Leong KH, Thomas NF, Chin HK, Ariffin A, Awang K. Resveratrol analogue, (E)-N-(2-(4-methoxystyryl) phenyl) furan-2-carboxamide induces G2/M cell cycle arrest through the activation of p53–p21CIP1/WAF1 in human colorectal HCT116 cells. Apoptosis 2018; 23:329-342. [DOI: 10.1007/s10495-018-1457-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
317
|
Maruyama K, Sheng Y, Watanabe H, Fukuzawa K, Tanaka S. Application of singular value decomposition to the inter-fragment interaction energy analysis for ligand screening. COMPUT THEOR CHEM 2018. [DOI: 10.1016/j.comptc.2018.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
318
|
Anticancer Profiling for Coumarins and Related O-Naphthoquinones from Mansonia gagei against Solid Tumor Cells In Vitro. Molecules 2018; 23:molecules23051020. [PMID: 29701706 PMCID: PMC6102575 DOI: 10.3390/molecules23051020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/17/2018] [Accepted: 04/21/2018] [Indexed: 11/17/2022] Open
Abstract
Napthoquinones and coumarins are naturally occurring compounds with potential anticancer activity. In the current study, two O-naphthoquinons (mansonone-G and mansonone-N) and six coumarins (mansorin-A, mansorin-B, mansorin-C, mansorins-I, mansorin-II, and mansorin-III) were isolated from the heartwood of Mansonia gagei family Sterculariaceae. Isolated compounds were examined for their potential anticancer activity against breast (MCF-7), cervix (HeLa), colorectal (HCT-116) and liver (HepG2) cancer cells using Sulfarhodamine-B (SRB) assay. Mansorin-II and mansorin-III showed relatively promising cytotoxic profile in all cell lines under investigation with inhibitory concentrations (IC50s) in the range of 0.74 µM to 36 µM and 3.95 µM to 35.3 µM, respectively. In addition, mansorin-B, mansorin-C, mansorin-II and mansorin-III significantly increased cellular entrapment of the P-glycoprotein (P-gp) substrate, doxorubicin, in colorectal cancer cells expressing the P-gp pump. The inhibitory effect of the isolated compounds on P-gp pump was examined using human recombinant P-gp molecules attached to ATPase subunit. Mansorin-B and mansonone-G were found to inhibit the P-gp attached ATPase subunit. On the other hand, mansorin-C, mansorin-III and mansorin-II inhibited P-gp pump via dual action (P-gp related ATPase subunit inhibition and P-gp substrate binding site occupation). However, mansorin II was examined for its potential chemomodulatory effect to paclitaxel (PTX) against colorectal cancer cells (HCT-116 and CaCo-2). Mansorin-II significantly reduced the IC50 of PTX in HCT-116 cells from 27.9 ± 10.2 nM to 5.1 ± 1.9 nM (synergism with combination index of 0.44). Additionally, Mansorin-II significantly reduced the IC50 of PTX in CaCo-2 cells from 2.1 ± 0.8 µM to 0.13 ± 0.03 µM (synergism with combination index of 0.18). Furthermore, cell cycle analysis was studied after combination of mansorin-II with paclitaxel using DNA flow cytometry analysis. Synergism of mansorin-II and PTX was reflected in increasing apoptotic cell population in both HCT-116 and CaCo-2 cells compared to PTX treatment alone. Combination of mansorin-II with PTX in CaCo-2 cells significantly increased the cell population in G2/M phase (from 2.9 ± 0.3% to 7.7 ± 0.8%) with reciprocal decrease in G0/G1 cell fraction from 52.1 ± 1.1% to 45.5 ± 1.0%. Similarly in HCT-116 cells, mansorin-II with PTX significantly increased the cell population in G2/M phase (from 33.4 ± 2.8% to 37.6 ± 1.3%) with reciprocal decrease in the S-phase cell population from 22.8 ± 1.7% to 20.2 ± 0.8%. In conclusion, mansorin-II synergizes the anticancer effect of paclitaxel in colorectal cancer cells, which might be partially attributed to enhancing its cellular entrapment via inhibiting P-gp efflux pump.
Collapse
|
319
|
Al-Anbaky Q, Al-Karakooly Z, Connor R, Williams L, Yarbrough A, Bush J, Ali N. Role of inositol polyphosphates in programed cell death in Dictyostelium discoideum and its developmental life cycle. Mol Cell Biochem 2018; 449:237-250. [PMID: 29679279 DOI: 10.1007/s11010-018-3360-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/16/2018] [Indexed: 11/28/2022]
Abstract
Programed cell death or apoptosis is a key developmental process that maintains tissue homeostasis in multicellular organisms. Inositol polyphosphates (InsPs) are key signaling molecules known to regulate a variety of cellular processes including apoptosis in such organisms. The signaling role of InsPs in unicellular organisms such as Dictyostelium discoideum (D. discoideum) is not well understood. We investigated whether InsPs also play any role in apoptosis in D. discoideum and whether InsPs-mediated apoptosis follows a mechanism similar to that present in higher multicellular eukaryotes. We measured known apoptotic markers in response to exogenously administered InsP6, the major InsPs in the cell. We found that InsP6 was able to cause cell death in D. discoideum cell culture in a dose- and time-dependent manner as determined by cytotoxicity assays. Fluorescence staining with acridine orange/ethidium bromide and flow cytometry results confirmed that the cell death in D. discoideum by InsP6 was due to apoptotic changes. Poly(ADP-ribose) expression, a known apoptotic marker used in D. discoideum, was also increased following InsP6 treatment suggesting a role for InsP6-mediated apoptosis in this organism. InsP6-mediated cell death was accompanied by production of reactive oxygen species and a decrease in mitochondrial membrane potential. Additionally, we studied the effects of InsP6 on the developmental life cycle of D. discoideum, the process likely affected by apoptosis. In conclusion, our studies provide evidence that InsP6-mediated cell death process is conserved in D. discoideum and plays an important signaling role in its developmental life cycle.
Collapse
Affiliation(s)
- Qudes Al-Anbaky
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA.,Department of Biology, University of Diyala, Baquba, Iraq
| | - Zeiyad Al-Karakooly
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Richard Connor
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Lisa Williams
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Azure Yarbrough
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - John Bush
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Nawab Ali
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR, 72204, USA.
| |
Collapse
|
320
|
Singh K, Gangrade A, Bhowmick S, Jana A, Mandal BB, Das N. Self-Assembly of a [1 + 1] Ionic Hexagonal Macrocycle and Its Antiproliferative Activity. Front Chem 2018; 6:87. [PMID: 29666793 PMCID: PMC5891631 DOI: 10.3389/fchem.2018.00087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/12/2018] [Indexed: 11/13/2022] Open
Abstract
A unique irregular hexagon was self-assembled using an organic donor clip (bearing terminal pyridyl units) and a complementary organometallic acceptor clip. The resulting metallamacrocycle was characterized by multinuclear NMR, mass spectrometry, and elemental analyses. Molecular modeling confirmed hexagonal shaped cavity for this metallamacrocycle which is a unique example of a discrete hexagonal framework self-assembled from only two building blocks. Cytotoxicity of the Pt-based acceptor tecton and the self-assembled PtII-based macrocycle was evaluated using three cancer cell lines and results were compared with cisplatin. Results confirmed a positive effect of the metallamacrocycle formation on cell growth inhibition.
Collapse
Affiliation(s)
- Khushwant Singh
- Department of Chemistry, Indian Institute of Technology Patna, Bihta, India
| | - Ankit Gangrade
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sourav Bhowmick
- Department of Chemistry, Indian Institute of Technology Patna, Bihta, India
| | - Achintya Jana
- Department of Chemistry, Indian Institute of Technology Patna, Bihta, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Neeladri Das
- Department of Chemistry, Indian Institute of Technology Patna, Bihta, India
| |
Collapse
|
321
|
Manafi Shabestari R, Alikarami F, Bashash D, Paridar M, Safa M. Overexpression of MiR-138 Inhibits Cell Growth and Induces Caspase-mediated Apoptosis in Acute Promyelocytic Leukemia Cell Line. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:24-31. [PMID: 30234070 PMCID: PMC6134423 DOI: 10.22088/ijmcm.bums.7.1.24] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 03/27/2018] [Indexed: 12/27/2022]
Abstract
Dysregulated expression of miRNAs can play a vital role in pathogenesis of leukemia. The shortened telomere length, and elevated telomerase activity in acute promyelocytic leukemia cells are mainly indicative of extensive proliferative activity. This study aimed to investigate the effect of overexpression of miR-138 on telomerase activity, and cell proliferation of acute promyelocytic leukemia NB4 cells. MiR-138 was overexpressed in NB4 cells using GFP hsa-miR-138-expressing lentiviruses. hTERT mRNA and protein expression levels were assessed by qRT-PCR and western blot analysis. For evaluation of apoptosis, annexin-V staining and activation of caspases were assessed using flow cytometry and western blot analysis, respectively. Our data demonstrate that overexpression of miR-138 attenuated the hTERT mRNA and protein expression levels. In addition, cell growth was inhibited, and malignant cells underwent caspase mediated-apoptosis in response to miR-138 overexpression. These findings suggest that loss of miR-138 expression may be associated with increased telomerase activity in NB4 cells. Therefore, strategies for up-regulation of miR-138 may result in inhibition of malignant cell growth, and provide a promising therapeutic approach for acute promyelocytic leukemia.
Collapse
Affiliation(s)
- Rima Manafi Shabestari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Alikarami
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Paridar
- Ministry of Health and Medical Education, Deputy of Management and Resources Development, Tehran, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
322
|
Petrilli AM, Garcia J, Bott M, Klingeman Plati S, Dinh CT, Bracho OR, Yan D, Zou B, Mittal R, Telischi FF, Liu XZ, Chang LS, Welling DB, Copik AJ, Fernández-Valle C. Ponatinib promotes a G1 cell-cycle arrest of merlin/NF2-deficient human schwann cells. Oncotarget 2018; 8:31666-31681. [PMID: 28427224 PMCID: PMC5458238 DOI: 10.18632/oncotarget.15912] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 02/20/2017] [Indexed: 02/04/2023] Open
Abstract
Neurofibromatosis type 2 (NF2) is a genetic syndrome that predisposes individuals to multiple benign tumors of the central and peripheral nervous systems, including vestibular schwannomas. Currently, there are no FDA approved drug therapies for NF2. Loss of function of merlin encoded by the NF2 tumor suppressor gene leads to activation of multiple mitogenic signaling cascades, including platelet-derived growth factor receptor (PDGFR) and SRC in Schwann cells. The goal of this study was to determine whether ponatinib, an FDA-approved ABL/SRC inhibitor, reduced proliferation and/or survival of merlin-deficient human Schwann cells (HSC). Merlin-deficient HSC had higher levels of phosphorylated PDGFRα/β, and SRC than merlin-expressing HSC. A similar phosphorylation pattern was observed in phospho-protein arrays of human vestibular schwannoma samples compared to normal HSC. Ponatinib reduced merlin-deficient HSC viability in a dose-dependent manner by decreasing phosphorylation of PDGFRα/β, AKT, p70S6K, MEK1/2, ERK1/2 and STAT3. These changes were associated with decreased cyclin D1 and increased p27Kip1levels, leading to a G1 cell-cycle arrest as assessed by Western blotting and flow cytometry. Ponatinib did not modulate ABL, SRC, focal adhesion kinase (FAK), or paxillin phosphorylation levels. These results suggest that ponatinib is a potential therapeutic agent for NF2-associated schwannomas and warrants further in vivo investigation.
Collapse
Affiliation(s)
- Alejandra M Petrilli
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| | - Jeanine Garcia
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| | - Marga Bott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| | - Stephani Klingeman Plati
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| | - Christine T Dinh
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Olena R Bracho
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Denise Yan
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Bing Zou
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Rahul Mittal
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Fred F Telischi
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Xue-Zhong Liu
- University of Miami Miller School of Medicine, Department of Otolaryngology, Miami, FL 33136, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - D Bradley Welling
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.,Current Affiliation: Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Massachusetts General Hospital and Harvard University, Boston, MA 02114, USA
| | - Alicja J Copik
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| | - Cristina Fernández-Valle
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Lake Nona-Orlando, FL 32827, USA
| |
Collapse
|
323
|
|
324
|
Griffin F, Marignol L. Therapeutic potential of melatonin for breast cancer radiation therapy patients. Int J Radiat Biol 2018. [PMID: 29521142 DOI: 10.1080/09553002.2018.1446227] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Melatonin is an endogenous hormone primarily known for its action on the circadian rhythms. But pre-clinical studies are reporting both its radioprotective and radiosensitizing properties, possibly mediated through an interaction between melatonin and the regulation of estrogens. Melatonin pre-treatment prior to ionizing radiation was associated with a decrease in cell proliferation and an increase in p53 mRNA expression, leading to an increase in the radiosensitivity of breast cancer cells. At the same time, a decrease in radiation-induced side effects was described in breast cancer patients and in rodent models. This review examines the potential for melatonin to improve the therapeutic outcomes of breast radiation therapy, specifically estrogen receptor positive patients. Evidence suggests that melatonin may offer a novel, non-toxic and cheap adjuvant therapy to improve the existing treatment modalities. But further research is required in the clinical setting before a clear understanding of its therapeutic benefits is determined.
Collapse
Affiliation(s)
- Fiona Griffin
- a Applied Radiation Therapy Trinity, Discipline of Radiation therapy , Trinity College Dublin , Dublin , Ireland
| | - Laure Marignol
- a Applied Radiation Therapy Trinity, Discipline of Radiation therapy , Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
325
|
Malojirao VH, Vigneshwaran V, Thirusangu P, Mahmood R, Prabhakar BT. The tumor antagonistic steroidal alkaloid Solanidine prompts the intrinsic suicidal signal mediated DFF-40 nuclear import and nucleosomal disruption. Life Sci 2018. [PMID: 29524520 DOI: 10.1016/j.lfs.2018.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aim Deformity in the cellular homeostatic event associated with cell survival and apoptosis are committing factors for carcinogenesis. Interventions of these events by pharmacologically active agent gain predominance in cancer treatment. In current investigation Solanidine, a steroidal alkaloid was evaluated on tumorigenesis by targeting death signal using multiple tumor cells and model systems. MAIN METHODS Anti-proliferative effect was evaluated using cytotoxic studies. Prolonged cytotoxic effect of Solanidine was examined by colony formation assay. Exhibition of apoptotic hallmark induced by Solanidine was examined using FACS analysis, Annexin-V staining, Acridine orange staining, TUNEL assay. Altered gene expression was evaluated using Immunoblot, Immunofluorescence and Immunohistochemistry technique. In-vitro results were revalidated in EAC solid tumor and CAM xenograft model. KEY FINDINGS Solanidine exerts its potential effect in a target specific manner. The cytotoxic/anticlonogenic activity was due to induction of typical cellular apoptotic hallmarks and cell cycle blockage at S-G2/M phase. The molecular events underlying this effect is through activation of intrinsic pathway via Bax, Bad and Cytochrome c activation by neutralizing Bcl-2 expression, along with downregulated PI3K/Akt survival signal. As a consequence, downstream pro apoptogenic gene, active Caspase-3 was over expressed by Solanidine to cleave its substrate PARP and promotes nuclear import of DFF-40. Anti-carcinogenic aptitude was further confirmed by murine solid tumors and in-vivo CAM xenograft studies. SIGNIFICANCE Solanidine emerged as active molecule against tomorigenesis by activating nuclear import of DFF-40 mediated nucleosomal disruption and cell demise. It can be developed as a potential apoptogenic small molecule for cancer therapy.
Collapse
Affiliation(s)
- Vikas H Malojirao
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga 577203, Karnataka, India
| | - V Vigneshwaran
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga 577203, Karnataka, India; Department of Pharmacology, Centre for Lung and Vascular Biology, University of Illinois at Chicago, Chicago 60612, USA
| | - Prabhu Thirusangu
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga 577203, Karnataka, India
| | - Riaz Mahmood
- Postgraduate Department of Studies and Research in Biotechnology & Bioinformatics, Kuvempu University, Shankaraghatta, Shivamogga 577203, Karnataka, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College (Autonomous), Kuvempu University, Shivamogga 577203, Karnataka, India.
| |
Collapse
|
326
|
Nikolian VC, Dennahy IS, Higgins GA, Williams AM, Weykamp M, Georgoff PE, Eidy H, Ghandour MH, Chang P, Alam HB. Transcriptomic changes following valproic acid treatment promote neurogenesis and minimize secondary brain injury. J Trauma Acute Care Surg 2018; 84:459-465. [PMID: 29251707 PMCID: PMC5905703 DOI: 10.1097/ta.0000000000001765] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Early treatment with valproic acid (VPA) has demonstrated benefit in preclinical models of traumatic brain injury, including smaller brain lesion size, decreased edema, reduced neurologic disability, and faster recovery. Mechanisms underlying these favorable outcomes are not fully understood. We hypothesized that VPA treatment would upregulate genes involved in cell survival and proliferation and downregulate those associated with cell death and the inflammatory response. METHODS Ten female swine were subjected to a protocol of traumatic brain injury and hemorrhagic shock. They were assigned to two groups (n = 5): normal saline (NS; 3× volume of shed blood), or NS + VPA (150 mg/kg). Following 6 hours of observation, brain tissue was harvested to evaluate lesion size and edema. Brain tissue was processed for RNA sequencing. Gene set enrichment and pathway analysis was performed to determine the differential gene expression patterns following injury. RESULTS Animals treated with VPA were noted to have a 46% reduction in brain lesion size and a 57% reduction in ipsilateral brain edema. Valproic acid significantly upregulated genes involved in morphology of the nervous system, neuronal development and neuron quantity. The VPA treatment downregulated pathways related to apoptosis, glial cell proliferation, and neuroepithelial cell differentiation. Ingenuity Pathway Analysis identified VPA as the top upstream regulator of activated transcription, supporting it as a direct cause of these transcriptional changes. Master transcriptional regulator NEUROD1 was also significantly upregulated, suggesting that VPA may induce additional transcription factors. CONCLUSION Administration of VPA attenuated brain lesion size, reduced brain edema, and induced significant changes in the transcriptome of injured brain within 6 hours. Patterns of differential expression were consistent with the proposed neurogenic and prosurvival effects of VPA treatment.
Collapse
Affiliation(s)
| | | | - Gerald A. Higgins
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI
| | | | - Michael Weykamp
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - Hassan Eidy
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - Panpan Chang
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Hasan B. Alam
- Department of Surgery, University of Michigan, Ann Arbor, MI
| |
Collapse
|
327
|
Trajano LADSN, Sergio LPDS, Stumbo AC, Mencalha AL, Fonseca ADSD. Low power lasers on genomic stability. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 180:186-197. [DOI: 10.1016/j.jphotobiol.2018.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/02/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
|
328
|
Wang W, Xu B, Li Q, Jiang D, Yan S. Anti‑cancer effects of a novel Pan‑RAF inhibitor in a hepatocellular carcinoma cell line. Mol Med Rep 2018; 17:6185-6193. [PMID: 29484394 DOI: 10.3892/mmr.2018.8615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 02/01/2018] [Indexed: 11/06/2022] Open
Abstract
The RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK (RAF/MEK/ERK) signaling cascade serves a prominent role in hepatocellular carcinoma (HCC) proliferation. Sorafenib (BAY 43‑9006) is a potent multikinase inhibitor of RAF kinases and a few receptor tyrosine kinases. Additionally, sorafenib causes apoptosis in a number of human tumor cell lines such as leukemia cell lines. Sorafenib is the first targeted drug to prolong the overall survival of patients with advanced HCC. However, sorafenib activity is less favorable in certain cancers, including sarcomas and melanomas, due to patient insensitivity and drug resistance. In the present study, a novel bi‑aryl urea, N‑(3‑trifluoromethylphenyl)‑N'-(2-methyl-4-(6‑cyclopropanecarboxamido-pyrimidin-4-yl) oxyphenyl) urea (CBI‑5725), is shown to be a potential candidate for the treatment of liver cancer. In the present study, the in vitro activities of CBI‑5725 and sorafenib in PLC/PRF/5 HCC cells were examined and the corresponding in vivo antitumor activities in PLC/PRF/5 human tumor xenografts. An alamar blue assay confirmed that CBI‑5725 was more cytotoxic than sorafenib to PLC/PRF/5 cells, suggesting that CBI‑5725 inhibited tumor cell proliferation more potently than sorafenib. CBI‑5725 inhibited the RAF/MEK/ERK signaling pathway to the same extent as sorafenib. In addition, CBI‑5725 elicited cell cycle arrest in the G2/M phase, while sorafenib did not markedly alter the cell cycle. Furthermore, CBI‑5725 induced apoptosis more strongly than sorafenib in a dose‑dependent manner, which may be attributed to greater caspase‑3 and poly(adenosine 5'‑diphosphate‑ribose) polymerase activation by CBI‑5725. In the PLC/PRF/5 xenograft model, 2 mg/kg CBI‑5725 inhibited tumor growth by 73%. At doses ranging from 6 to 18 mg/kg, CBI‑5725 nearly completely prevented tumor growth. These results imply that the antitumor efficacy of CBI‑5725 in HCC models may result from the suppression of the RAF/MEK/ERK signaling pathway, the induction of cell cycle arrest in the G2/M phase, and the initiation of caspase‑3‑dependent apoptosis. These observations suggested that CBI‑5725 may be a potent novel compound for the treatment of HCC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Qixiang Li
- Biomarker and Diagnostic Technology, Crown Bioscience Inc., Beijing 102200, P.R. China
| | - Dechun Jiang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Suying Yan
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
329
|
Sergio LPS, Lucinda LMF, Reboredo MM, de Paoli F, Fonseca LMC, Pinheiro BV, Mencalha AL, Fonseca AS. Emphysema induced by elastase alters the mRNA relative levels from DNA repair genes in acute lung injury in response to sepsis induced by lipopolysaccharide administration in Wistar rats. Exp Lung Res 2018; 44:79-88. [DOI: 10.1080/01902148.2017.1422158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Luiz Philippe S. Sergio
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
| | - Leda M. F. Lucinda
- Laboratório de Pesquisa em Pneumologia, Universidade Federal de Juiz de Fora, Dom Bosco, Juiz de Fora, Minas Gerais, Brazil
- Centro de Biologia da Reprodução, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Maycon M. Reboredo
- Laboratório de Pesquisa em Pneumologia, Universidade Federal de Juiz de Fora, Dom Bosco, Juiz de Fora, Minas Gerais, Brazil
- Centro de Biologia da Reprodução, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Flavia de Paoli
- Centro de Biologia da Reprodução, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Lídia M. C. Fonseca
- Laboratório de Pesquisa em Pneumologia, Universidade Federal de Juiz de Fora, Dom Bosco, Juiz de Fora, Minas Gerais, Brazil
- Centro de Biologia da Reprodução, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Bruno V. Pinheiro
- Laboratório de Pesquisa em Pneumologia, Universidade Federal de Juiz de Fora, Dom Bosco, Juiz de Fora, Minas Gerais, Brazil
- Centro de Biologia da Reprodução, Universidade Federal de Juiz de Fora, Campus Universitário, São Pedro, Juiz de Fora, Minas Gerais, Brazil
| | - Andre L. Mencalha
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
| | - Adenilson S. Fonseca
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Rio de Janeiro, Brazil
- Centro de Ciências da Saúde, Centro Universitário Serra dos Órgãos, Teresópolis, Rio de Janeiro, Brazil
| |
Collapse
|
330
|
Han YH, Kee JY, Hong SH. Rosmarinic Acid Activates AMPK to Inhibit Metastasis of Colorectal Cancer. Front Pharmacol 2018; 9:68. [PMID: 29459827 PMCID: PMC5807338 DOI: 10.3389/fphar.2018.00068] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/18/2018] [Indexed: 12/16/2022] Open
Abstract
Rosmarinic acid (RA) has been used as an anti-inflammatory, anti-diabetic, and anti-cancer agent. Although RA has also been shown to exert an anti-metastatic effect, the mechanism of this effect has not been reported to be associated with AMP-activated protein kinase (AMPK). The aim of this study was to elucidate whether RA could inhibit the metastatic properties of colorectal cancer (CRC) cells via the phosphorylation of AMPK. RA inhibited the proliferation of CRC cells through the induction of cell cycle arrest and apoptosis. In several metastatic phenotypes of CRC cells, RA regulated epithelial-mesenchymal transition (EMT) through the upregulation of an epithelial marker, E-cadherin, and the downregulation of the mesenchymal markers, N-cadherin, snail, twist, vimentin, and slug. Invasion and migration of CRC cells were inhibited and expressions of matrix metalloproteinase (MMP)-2 and MMP-9 were decreased by RA treatment. Adhesion and adhesion molecules such as ICAM-1 and integrin β1 expressions were also reduced by RA treatment. In particular, the effects of RA on EMT and MMPs expressions were due to the activation of AMPK. Moreover, RA inhibited lung metastasis of CRC cells by activating AMPK in mouse model. Collectively, these results proved that RA could be potential therapeutic agent against metastasis of CRC.
Collapse
Affiliation(s)
- Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| |
Collapse
|
331
|
Chang YW, Singh KP. Duration-dependent effects of nicotine exposure on growth and AKT activation in human kidney epithelial cells. Mol Cell Biochem 2018; 448:51-60. [PMID: 29396723 DOI: 10.1007/s11010-018-3312-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/27/2018] [Indexed: 01/06/2023]
Abstract
Exposure to nicotine is known to cause adverse effects in many target organs including kidney. Epidemiological studies suggest that nicotine-induced kidney diseases are prevalent worldwide. However, the impact of duration of exposure on the nicotine-induced adverse effects in normal kidney cells and the underlying molecular mechanism is still unclear. Hence, the objective of this study was to evaluate both acute and long-term effects of nicotine in normal human kidney epithelial cells (HK-2). Cells were treated with 1 and 10 µM nicotine for acute and long-term duration. The result of cell viability showed that the acute exposure to 1 µM nicotine has no significant effect on growth. However, the 10 µM nicotine caused significant decrease in the growth of HK-2 cells. The long-term exposure resulted in significantly increased cell growth in both 1 and 10 µM nicotine-treated groups. Analysis of cell cycle and expression of marker genes related to proliferation and apoptosis further confirmed the effects of nicotine. Additionally, the analysis of growth signaling pathway revealed the decreased level of pAKT in cells with acute exposure whereas the increased level of pAKT in long-term nicotine-exposed cells. This suggests that nicotine, through modulating the AKT pathway, controls the duration-dependent effects on the growth of HK-2 cells. In summary, this is the first report showing long-duration exposure to nicotine causes increased proliferation of human kidney epithelial cells through activation of AKT pathway.
Collapse
Affiliation(s)
- Yu-Wei Chang
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, TX, 79409, USA
| | - Kamaleshwar P Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, TX, 79409, USA.
| |
Collapse
|
332
|
Novel enmein-type diterpenoid hybrids coupled with nitrogen mustards: Synthesis of promising candidates for anticancer therapeutics. Eur J Med Chem 2018; 146:588-598. [DOI: 10.1016/j.ejmech.2018.01.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022]
|
333
|
Abu N, Zamberi NR, Yeap SK, Nordin N, Mohamad NE, Romli MF, Rasol NE, Subramani T, Ismail NH, Alitheen NB. Subchronic toxicity, immunoregulation and anti-breast tumor effect of Nordamnacantal, an anthraquinone extracted from the stems of Morinda citrifolia L. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:31. [PMID: 29374471 PMCID: PMC5787285 DOI: 10.1186/s12906-018-2102-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/17/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Morinda citrifolia L. that was reported with immunomodulating and cytotoxic effects has been traditionally used to treat multiple illnesses including cancer. An anthraquinone derived from fruits of Morinda citrifolia L., nordamnacanthal, is a promising agent possessing several in vitro biological activities. However, the in vivo anti-tumor effects and the safety profile of nordamnacanthal are yet to be evaluated. METHODS In vitro cytotoxicity of nordamnacanthal was tested using MTT, cell cycle and Annexin V/PI assays on human MCF-7 and MDA-MB231 breast cancer cells. Mice were orally fed with nordamnacanthal daily for 28 days for oral subchronic toxicity study. Then, the in vivo anti-tumor effect was evaluated on 4T1 murine cancer cells-challenged mice. Changes of tumor size and immune parameters were evaluated on the untreated and nordamnacanthal treated mice. RESULTS Nordamnacanthal was found to possess cytotoxic effects on MDA-MB231, MCF-7 and 4T1 cells in vitro. Moreover, based on the cell cycle and Annexin V results, nordamnacanthal managed to induce cell death in both MDA-MB231 and MCF-7 cells. Additionally, no mortality, signs of toxicity and changes of serum liver profile were observed in nordamnacanthal treated mice in the subchronic toxicity study. Furthermore, 50 mg/kg body weight of nordamncanthal successfully delayed the progression of 4T1 tumors in Balb/C mice after 28 days of treatment. Treatment with nordamnacanthal was also able to increase tumor immunity as evidenced by the immunophenotyping of the spleen and YAC-1 cytotoxicity assays. CONCLUSION Nordamnacanthal managed to inhibit the growth and induce cell death in MDA-MB231 and MCF-7 cell lines in vitro and cease the tumor progression of 4T1 cells in vivo. Overall, nordamnacanthal holds interesting anti-cancer properties that can be further explored.
Collapse
|
334
|
Joksimović N, Baskić D, Popović S, Zarić M, Kosanić M, Ranković B, Stanojković T, Novaković SB, Davidović G, Bugarčić Z, Janković N. Synthesis, characterization, biological activity, DNA and BSA binding study: novel copper(ii) complexes with 2-hydroxy-4-aryl-4-oxo-2-butenoate. Dalton Trans 2018; 45:15067-15077. [PMID: 27711668 DOI: 10.1039/c6dt02257j] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A serie of novel square pyramidal copper(ii) complexes [Cu(L)2H2O] (3a-d) with O,O-bidentate ligands [L = ethyl-2-hydroxy-4-aryl-4-oxo-2-butenoate; aryl = 3-methoxyphenyl-2a, (E)-2-phenylvinyl-2b, (E)-2-(4'-hydroxy-3'-methoxyphenyl)vinyl-2c, 3-nitrophenyl-2d, 2-thienyl-2e] were synthesized and characterized by spectral (UV-Vis, IR, ESI-MS and EPR), elemental and X-ray analysis. The antimicrobial activity was estimated by the determination of the minimal inhibitory concentration (MIC) using the broth microdilution method. The most active antibacterial compounds were 3c and 3d, while the best antifungal activity was showed by complexes 3b and 3e. The lowest MIC value (0.048 mg mL-1) was measured for 3c against Proteus mirabilis. The cytotoxic activity was tested using the MTT method on human epithelial carcinoma HeLa cells, human lung carcinoma A549 cells and human colon carcinoma LS174 cells. All complexes showed extremely better cytotoxic activity compared to cisplatin at all tested concentrations. Compound 3d expressed the best activity against all tested cell lines with IC50 values ranging from 7.45 to 7.91 μg mL-1. The type of cell death and the impact on the cell cycle for 3d and 3e were evaluated by flow cytometry. Both compounds induced apoptosis and S phase cell cycle arrest. The interactions between selected complexes (3d and 3e) and CT-DNA or bovine serum albumin (BSA) were investigated by the fluorescence spectroscopic method. Competitive experiments with ethidium bromide (EB) indicated that 3d and 3e have a propensity to displace EB from the EB-DNA complex through intercalation suggesting strong competition with EB [Ksv = (1.4 ± 0.2) and (2.9 ± 0.1) × 104 M-1, respectively]. Ksv values indicate that these complexes bind to DNA covalently and non-covalently. The achieved results in the fluorescence titration of BSA with 3d and 3e [Ka = (2.9 ± 0.2) × 106 and (2.5 ± 0.2) × 105 M, respectively] showed that the fluorescence quenching of BSA is a result of the formation of the 3d- and 3e-BSA complexes. The obtained Ka values are high enough to ensure that a significant amount of 3d and 3e gets transported and distributed through the cells.
Collapse
Affiliation(s)
- Nenad Joksimović
- Faculty of Science, University of Kragujevac, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Dejan Baskić
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Suzana Popović
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Milan Zarić
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Marijana Kosanić
- Faculty of Science, University of Kragujevac, Department of Biology and Ecology, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Branislav Ranković
- Faculty of Science, University of Kragujevac, Department of Biology and Ecology, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Tatjana Stanojković
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Sladjana B Novaković
- Vinča Institute of Nuclear Science, University of Belgrade University of Belgrade, P.O. Box 522, 11001, Belgrade, Serbia
| | - Goran Davidović
- Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Zorica Bugarčić
- Faculty of Science, University of Kragujevac, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Nenad Janković
- Faculty of Science, University of Kragujevac, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| |
Collapse
|
335
|
Manickam V, Dhakshinamoorthy V, Perumal E. Iron Oxide Nanoparticles Induces Cell Cycle-Dependent Neuronal Apoptosis in Mice. J Mol Neurosci 2018; 64:352-362. [PMID: 29368134 DOI: 10.1007/s12031-018-1030-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/11/2018] [Indexed: 01/09/2023]
Abstract
Iron oxide (Fe2O3) nanoparticles (NPs) with its unique magnetic and paramagnetic properties are popular in biomedical applications. Some of their neurotoxic mechanisms due to repeated administration are proven. However, we speculate that the neuronal damage might be due to apoptosis resulting from unusual cell cycle entry. Moreover, iron accumulation has been shown to be closely associated with most of the neurodegenerative disorders. Thus, in the current study, mice were orally (po) treated with the Fe2O3-NPs to investigate cell cycle-associated events/components and occurrence of apoptosis. A subsequent increase in oxidant levels was observed with the iron accumulation due to Fe2O3-NPs exposure. The accumulated β-amyloid and reduced level of cdk5 seem to aid in the cell cycle entry and forcing progression towards apoptosis. Expression of Cyclin D1 and pRb (Ser 795) indicate the cell cycle re-entry of neurons. Overexpression of RNA Pol II and PARP cleavage suggests DNA damage due to Fe2O3-NPs exposure. Further, hyperphosphorylation of p38 (Thr 180/Tyr 182) confirms the activation of DNA damage-dependent checkpoint. Expression patterns of pro- and anti-apoptotic markers, TUNEL and TEM indicate the occurrences of apoptosis.
Collapse
Affiliation(s)
- Vijayprakash Manickam
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India
| | - Vasanth Dhakshinamoorthy
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, 641 046, India.
| |
Collapse
|
336
|
Spiclomazine displays a preferential anti-tumor activity in mutant KRas-driven pancreatic cancer. Oncotarget 2018; 9:6938-6951. [PMID: 29467941 PMCID: PMC5805527 DOI: 10.18632/oncotarget.24025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023] Open
Abstract
Ras-targeted therapy represents a 'holy grail' in oncology. Based on our model prediction, Spiclomazine freezing the intermediate conformation of activated Ras is central to cancer therapeutics. We show here that Spiclomazine leads to an effective suppression in Ras-mediated signaling through abrogating the KRas-GTP level in the KRas-driven pancreatic cancer. The Ras-mediated signaling inhibition leads to dramatically reduced survivals of five KRas-driven pancreatic cancer cell lines with IC50 ranging 19.7~74.2 μM after 48 hours of treatment. However, no significant changes have been observed for normal cell lines. It is worth mentioning that the mutant KRas-driven cancer cells are more sensitive towards Spiclomazine than the wild-type KRas cancer cells. Subsequent cellular thermal shift and RNA interference assays show that Spiclomazine efficiently binds with and stabilizes KRas to a certain extent within the cells. This validates the effect of target engagement on drug efficacy. Furthermore, Spiclomazine arrests cell cycle at G2 phase in the cancer cells, without obvious cell-cycle arrest in the normal cells. This further demonstrates its selectively biological response to cancer cells involved in Ras-GTP-mediated target engagement. Spiclomazine completely inhibits the growth of MIA PaCa-2 tumors on renal capsule xenograft models in BALB/c mice administered 68 mg kg-1 for 2 weeks via intra-peritoneal route. Immunohistochemical analyses reveal the reduced c-Raf and p-ERK and the increase in TUNEL staining. These observations further confirm the in vitro findings. Taken together, Spiclomazine is a selective inhibitor for mutant KRas-driven pancreatic cancer.
Collapse
|
337
|
Protein neddylation and its alterations in human cancers for targeted therapy. Cell Signal 2018; 44:92-102. [PMID: 29331584 DOI: 10.1016/j.cellsig.2018.01.009] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 01/05/2023]
Abstract
Neddylation, a post-translational modification that conjugates an ubiquitin-like protein NEDD8 to substrate proteins, is an important biochemical process that regulates protein function. The best-characterized substrates of neddylation are the cullin subunits of Cullin-RING ligases (CRLs), which, as the largest family of E3 ubiquitin ligases, control many important biological processes, including tumorigenesis, through promoting ubiquitylation and subsequent degradation of a variety of key regulatory proteins. Recently, increasing pieces of experimental evidence strongly indicate that the process of protein neddylation modification is elevated in multiple human cancers, providing sound rationale for its targeting as an attractive anticancer therapeutic strategy. Indeed, neddylation inactivation by MLN4924 (also known as pevonedistat), a small molecule inhibitor of E1 NEDD8-activating enzyme currently in phase I/II clinical trials, exerts significant anticancer effects by inducing cell cycle arrest, apoptosis, senescence and autophagy in a cell-type and context dependent manner. Here, we summarize the latest progresses in the field with a major focus on preclinical studies in validation of neddylation modification as a promising anticancer target.
Collapse
|
338
|
Paul P, Chatterjee S, Pramanik A, Karmakar P, Chandra Bhattacharyya S, Kumar GS. Thionine Conjugated Gold Nanoparticles Trigger Apoptotic Activity Toward HepG2 Cancer Cell Line. ACS Biomater Sci Eng 2018; 4:635-646. [DOI: 10.1021/acsbiomaterials.7b00390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Puja Paul
- Organic
and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
- Department
of Chemistry, Jadavpur University, Kolkata 700 032, India
| | - Sabyasachi Chatterjee
- Organic
and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| | - Arindam Pramanik
- Department
of Life Science and Bio-technology, Jadavpur University, Kolkata 700 032, India
| | - Parimal Karmakar
- Department
of Life Science and Bio-technology, Jadavpur University, Kolkata 700 032, India
| | | | - Gopinatha Suresh Kumar
- Organic
and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700 032, India
| |
Collapse
|
339
|
Lamy S, Muhire É, Annabi B. Antiproliferative efficacy of elderberries and elderflowers (Sambucus canadensis) on glioma and brain endothelial cells under normoxic and hypoxic conditions. J Funct Foods 2018; 40:164-179. [DOI: 10.1016/j.jff.2017.10.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
340
|
Abstract
High content imaging-based cell cycle analysis allows multiplexing of various parameters including DNA content, DNA synthesis, cell proliferation, and other cell cycle markers such as phosho-histone H3. 5'-Ethynyl-2'-deoxyuridine (EdU) incorporation is a thymidine analog that provides a sensitive method for the detection of DNA synthesis in proliferating cells that is a more convenient method than the traditional BrdU detection by antibody. Caspase 3 is activated in programmed cell death induced by both intrinsic (mitochondrial) and extrinsic factors (death ligand). Cell cycle and apoptosis are common parameters studied in the phenotypic analysis of compound toxicity and anti-cancer drugs. In this chapter, we describe methods for the detection of s-phase cell cycle progression by EdU incorporation, and caspase 3 activation using the CellEvent caspase 3/7 detection reagent.
Collapse
|
341
|
Desethylamiodarone-A metabolite of amiodarone-Induces apoptosis on T24 human bladder cancer cells via multiple pathways. PLoS One 2017; 12:e0189470. [PMID: 29220397 PMCID: PMC5722307 DOI: 10.1371/journal.pone.0189470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022] Open
Abstract
Bladder cancer (BC) is a common malignancy of the urinary tract that has a higher frequency in men than in women. Cytostatic resistance and metastasis formation are significant risk factors in BC therapy; therefore, there is great interest in overcoming drug resistance and in initiating research for novel chemotherapeutic approaches. Here, we suggest that desethylamiodarone (DEA)–a metabolite of amiodarone—may have cytostatic potential. DEA activates the collapse of mitochondrial membrane potential (detected by JC-1 fluorescence), and induces cell death in T24 human transitional-cell bladder carcinoma cell line at physiologically achievable concentrations. DEA induces cell cycle arrest in the G0/G1 phase, which may contribute to the inhibition of cell proliferation, and shifts the Bax/Bcl-2 ratio to initiate apoptosis, induce AIF nuclear translocation, and activate PARP-1 cleavage and caspase-3 activation. The major cytoprotective kinases—ERK and Akt—are inhibited by DEA, which may contribute to its cell death-inducing effects. DEA also inhibits the expression of B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1) and reduces colony formation of T24 bladder carcinoma cells, indicating its possible inhibitory effect on metastatic potential. These data show that DEA is a novel anti-cancer candidate of multiple cell death-inducing effects and metastatic potential. Our findings recommend further evaluation of its effects in clinical studies.
Collapse
|
342
|
Nam Dia long, a Vietnamese folk formula, induces apoptosis in MCF-7 cells through various mechanisms of action. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:522. [PMID: 29202775 PMCID: PMC5716261 DOI: 10.1186/s12906-017-2027-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022]
Abstract
Background The holistic approach of traditional medicine renders the identification of its mechanisms of action difficult. Microarray technology provides an efficient way to analyze the complex genome-wide gene expression of cells treated with mixtures of medicinal ingredients. We performed transcriptional profiling of MCF-7 cells treated with Nam Dia Long (NDL), a Vietnamese traditional formula, to explore the mechanism of action underlying the apoptosis inducing effect of this formula reported in a previous study. Methods MCF-7 cells were treated with aqueous extracts of NDL at the IC50 concentration for 24, 36 and 48 h. Total RNAs at 24 h and 48 h were subsequently extracted, reverse transcribed and submitted to microarray expression profiling using the Human HT-12 v4.0 Expression Bead Chip (Illumina). Functional analyses were performed using the Database for Annotation, Visualization and Integrated Discovery and the Ingenuity Pathways Analysis. The expression level from selected genes at the three time points were assessed by quantitative real-time RT-PCR and Western blot. Results Fifty-four and 601 genes were differentially expressed at 24 and 48 h of NDL treatment, respectively. Genes with altered expression at 24 h were mostly involved in cell responses to xenobiotic stress whereas genes differentially expressed at 48 h were related to endoplasmic reticulum stress, DNA damage and cell cycle control. Apoptosis of NDL treated MCF-7 cells resulted from a combination of different mechanisms including the intrinsic and extrinsic pathways, cell cycle arrest- and oxidative stress-related cell death. Conclusion NDL elicited a two-stage response in MCF-7 treated cells with apoptosis as the ultimate result. The various mechanisms inducing apoptosis reflected the complexity of the formula composition. Electronic supplementary material The online version of this article (10.1186/s12906-017-2027-2) contains supplementary material, which is available to authorized users.
Collapse
|
343
|
Yaftian M, Yari F, Ghasemzadeh M, Fallah Azad V, Haghighi M. Induction of Apoptosis in Cancer Cells of pre-B ALL Patients after Exposure to Platelets, Platelet-Derived Microparticles and Soluble CD40 Ligand. CELL JOURNAL 2017; 20:120-126. [PMID: 29308628 PMCID: PMC5759674 DOI: 10.22074/cellj.2018.5032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/12/2017] [Indexed: 11/13/2022]
Abstract
Objective The in vitro treatment of tumor cells with platelet (Plt) causes inhibition of tumor cell growth, although
mechanism of this effect is not clear yet. Induction of apoptosis has been proposed as a mechanism of Plt effects on
tumor cells. The purpose of this study was to clarify the role of Plts and Plt-derived components in the induction of
apoptosis in the blood mononuclear cells of patients with leukemia.
Materials and Methods In this experimental study, peripheral blood mononuclear cells (PBMCs) were isolated from
whole blood of five patients with childhood B-precursor acute lymphoblastic leukemia (pre-B ALL) and encountered with
Plts, Plt-derived microparticles (Plt-MPs) as well as purified soluble CD40L (sCD40L). After 48 hours of co-culture, the
anti-cancer activity of the aforementioned factors was surveyed using examination of apoptosis markers of the cells
including active caspase-3 and CD95 using ELISA and flow cytometer techniques, respectively. Additionally, staining of
the cells with 7-Aminoactinomycin D (7-AAD) was evaluated by flow cytometer technique. Trypan blue exclusion test
and WST-1 method were also used to compare the death/survival status of the cells.
Results Levels of CD95 and caspase-3 were significantly increased in the all treated groups (P<0.05). On the other
hand, trypan blue, 7-AAD and WST-1 methods showed significantly lower number of the live cells in the treated groups
(P<0.05).
Conclusion This study can show the ability of Plts, Plt-MPs and sCD40L for the induction of apoptosis in PBMCs of
pre-B-ALL patients. Further studies are necessary to elucidate the different effects of platelets on cancer cells in vitro
and in vivo.
Collapse
Affiliation(s)
- Morteza Yaftian
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Yari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran. Electronic address :
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | | | |
Collapse
|
344
|
Li P, Fei H, Wang L, Xu H, Zhang H, Zheng L. PDCD5 regulates cell proliferation, cell cycle progression and apoptosis. Oncol Lett 2017; 15:1177-1183. [PMID: 29403562 DOI: 10.3892/ol.2017.7401] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/03/2017] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death (PDCD)5 is cloned from human leukemia cell line TF-1. PDCD5 is one of the members of the programmed cell death protein family that is frequently involved in tumor growth and apoptosis. To investigate the molecular and cellular functions of PDCD5, the present study established a PDCD5 stably overexpressing A431 cell line and examined the role of PDCD5 in cell proliferation, cell cycle progression and apoptosis. The data demonstrated that overexpression of PDCD5 significantly inhibited cell proliferation, induced cell cycle arrest at G2/M phase and apoptosis in A431 cells. The expression profiles of certain key regulators of these cellular events were further investigated, including P53, B cell lymphoma (BCL)-2, BCL-2 associated X protein (BAX) and caspase (CASP)3. The data demonstrated that at the transcript and protein levels, P53, BAX and CASP3 were all upregulated in the PDCD5 stably overexpressing A431 cells whereas BCL-2 was downregulated, indicating that PDCD5 acts as an important upstream regulator of P53, BCL-2, BAX and CASP3. The data suggest that PDCD5 regulates cell proliferation, cell cycle progression and apoptosis in A431 cells. PDCD5 may be a novel tumor suppressor gene, and may be potentially used for cancer treatment in the future.
Collapse
Affiliation(s)
- Penghui Li
- Department of Biogenetics, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Hongxin Fei
- Department of Histology and Embryology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Lihong Wang
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Huiyu Xu
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haiyan Zhang
- Department of Histology and Embryology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Lihong Zheng
- Department of Biogenetics, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
345
|
Ji W, Liu S, Zhao X, Guo Y, Xia S, Lu Y, Yin M, Xu X. Triptolide inhibits proliferation, differentiation and induces apoptosis of osteoblastic MC3T3‑E1 cells. Mol Med Rep 2017; 16:7391-7397. [PMID: 28944904 PMCID: PMC5865870 DOI: 10.3892/mmr.2017.7568] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 04/10/2017] [Indexed: 01/16/2023] Open
Abstract
Ankylosing spondylitis (AS) is characterized by the formation of bony spurs. Treatment of the resulting ankylosis, excessive bone formation and associated functional impairment, remain the primary therapeutic aims in research regarding this condition. Triptolide is the primary active component of the perennial vine Tripterygium wilfordii Hook. f., and has previously been demonstrated to exert anti‑tumor activities including inhibition of cell growth and the induction of apoptosis, however, the effect of triptolide on osteoblasts remains to be elucidated. In the present study, the MC3T3‑E1 mouse osteoblast cell line was treated with differing concentrations of triptolide for various intervals. Cell proliferation was detected using the bromodeoxyuridine assay, cell cycle and apoptosis were measured by flow cytometry, nuclear apoptosis was observed by Hoechst staining and associated proteins were determined via western blot analysis. The cells were then further incubated with osteogenic induction medium supplemented with triptolide for 7 or 12 days and the differentiation to osteoblasts was examined by picrosirius staining, observation of alkaline phosphatase activity and a calcium deposition assay. It was demonstrated that treatment with triptolide significantly inhibited osteoblast proliferation and induced cell cycle arrest and apoptosis of the osteoblasts. Furthermore, treatment with triptolide reduced collagen formation, alkaline phosphatase activity and calcium deposition. The present study demonstrated an inhibitory effect of triptolide on osteoblast proliferation and differentiation, and therefore suggests a potential therapeutic agent for the treatment of AS in the future.
Collapse
Affiliation(s)
- Wei Ji
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Shijia Liu
- Laboratory of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Xia Zhao
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Yunke Guo
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Sha Xia
- Hematology-Oncology Department, Zhenjiang Hospital of Integrated Chinese and Western Medicine, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yueyang Lu
- College of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Menyun Yin
- Hematology-Oncology Department, Zhenjiang Hospital of Integrated Chinese and Western Medicine, Zhenjiang, Jiangsu 212000, P.R. China
| | - Xiao Xu
- College of Nursing, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
346
|
Ryu JM, Jang GY, Woo KS, Kim TM, Jeong HS, Kim DJ. Effects of sorghum ethyl-acetate extract on PC3M prostate cancer cell tumorigenicity. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
347
|
Al-Maleki AR, Loke MF, Lui SY, Ramli NSK, Khosravi Y, Ng CG, Venkatraman G, Goh KL, Ho B, Vadivelu J. Helicobacter pylori outer inflammatory protein A (OipA) suppresses apoptosis of AGS gastric cells in vitro. Cell Microbiol 2017; 19. [PMID: 28776327 DOI: 10.1111/cmi.12771] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 07/02/2017] [Accepted: 07/26/2017] [Indexed: 12/12/2022]
Abstract
Outer inflammatory protein A (OipA) is an important virulence factor associated with gastric cancer and ulcer development; however, the results have not been well established and turned out to be controversial. This study aims to elucidate the role of OipA in Helicobacter pylori infection using clinical strains harbouring oipA "on" and "off" motifs. Proteomics analysis was performed on AGS cell pre-infection and postinfection with H. pylori oipA "on" and "off" strains, using liquid chromatography/mass spectrometry. AGS apoptosis and cell cycle assays were performed. Moreover, expression of vacuolating cytotoxin A (VacA) was screened using Western blotting. AGS proteins that have been suggested previously to play a role or associated with gastric disease were down-regulated postinfection with oipA "off" strains comparing to oipA "on" strains. Furthermore, oipA "off" and ΔoipA cause higher level of AGS cells apoptosis and G0/G1 cell-cycle arrest than oipA "on" strains. Interestingly, deletion of oipA increased bacterial VacA production. The capability of H. pylori to induce apoptosis and suppress expression of proteins having roles in human disease in the absence of oipA suggests that strains not expressing OipA may be less virulent or may even be protective against carcinogenesis compared those expressing OipA. This potentially explains the higher incidence of gastric cancer in East Asia where oipA "on" strains predominates.
Collapse
Affiliation(s)
- Anis Rageh Al-Maleki
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mun Fai Loke
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sook Yin Lui
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nur Siti Khadijah Ramli
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yalda Khosravi
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chow Goon Ng
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gopinath Venkatraman
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Khean-Lee Goh
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Bow Ho
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Precision Medicine Centre Pte Ltd, Singapore, Singapore
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
348
|
Navanesan S, Abdul Wahab N, Manickam S, Cheow YL, Sim KS. Intrinsic capabilities of Leptospermum javanicum in inducing apoptosis and suppressing the metastatic potential of human lung carcinoma cells. Chem Biol Interact 2017; 273:37-47. [DOI: 10.1016/j.cbi.2017.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/14/2017] [Accepted: 05/30/2017] [Indexed: 10/19/2022]
|
349
|
Čanović P, Simović AR, Radisavljević S, Bratsos I, Demitri N, Mitrović M, Zelen I, Bugarčić ŽD. Impact of aromaticity on anticancer activity of polypyridyl ruthenium(II) complexes: synthesis, structure, DNA/protein binding, lipophilicity and anticancer activity. J Biol Inorg Chem 2017; 22:1007-1028. [DOI: 10.1007/s00775-017-1479-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/24/2017] [Indexed: 12/12/2022]
|
350
|
Lee C, Chun W, Zhao R, Kim YD, Nam MM, Jung DH, Cho IJ, Jegal KH, Lee TH, Kim YW, Park SM, Ju SA, Lee CW, Kim SC, An WG. Anticancer effects of an extract from the scallop Patinopecten yessoensis on MCF-7 human breast carcinoma cells. Oncol Lett 2017; 14:2207-2217. [PMID: 28789443 PMCID: PMC5530092 DOI: 10.3892/ol.2017.6424] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/03/2017] [Indexed: 01/15/2023] Open
Abstract
Patinopecten yessoensis, is a species of scallop and a marine bivalve mollusk. In traditional East Asian medicine, scallop meat is used as a drug for the treatment of diabetes, pollakisuria, and indigestion. The present study was conducted in order to examine the potential anticancer effects of scallop flesh extract (SE) on MCF-7 human breast cancer cells. An MTT assay was used to evaluate cell viability and flow cytometry was used for the assessment of cell cycle distribution and apoptosis. The alteration in protein expression level was determined by western blot analysis, and the amounts of docosahexaenoic acid and eicosapentaenoic acid in the SE were measured by gas chromatography. SE inhibited the growth of MCF-7 human breast cancer cells in a dose-dependent manner by inducing G0/G1 phase arrest. The cell cycle arrest was associated with the upregulation of p53 and p21, and downregulation of G1 phase-associated cyclin D1/cyclin-dependent kinase (Cdk) 4 and cyclin E1/Cdk 2. In addition, SE-mediated cell cycle arrest was associated with the promotion of apoptosis, as indicated by the expression of apoptosis-associated proteins and changes in nuclear morphology. SE appeared to induce the mitochondrial apoptotic cascade, as indicated by a decreased expression of Bcl-2, activation of Bcl-2 associated X protein, release of cytochrome c, decrease in procaspase-3, and an increase in cleaved-poly (ADP-ribose) polymerase (PARP). Furthermore, the expression levels of Fas-associated via death domain and cleaved caspase-8 were increased in a SE dose-dependent manner. Taken together, these results suggest that the intrinsic and extrinsic pathways of apoptosis are associated with the anticancer effects of SE on MCF-7 cells. Thus, SE may be a suitable candidate for the treatment and prevention of human breast cancer.
Collapse
Affiliation(s)
- Chu Lee
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Wonjoo Chun
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea
| | - Rongjie Zhao
- School of Mental Health, Qiqihar Medical University, Qiqihar, Heilongjiang 161042, P.R. China
| | - Young Dae Kim
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Myung Mo Nam
- Aquaculture Industry Division, NFRDI, Gangneung 210-809, Republic of Korea
| | - Dae Hwa Jung
- HaniBio Co., Ltd., Gyeongsan 712-260, Republic of Korea
| | - Il Je Cho
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Kyung Hwan Jegal
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Tae Hoon Lee
- Department of Biological Sciences, College of Biomedical Sciences and Engineering, Inje University, Gimhae 621-749, Republic of Korea
| | - Young Woo Kim
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Sang Mi Park
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Seong A Ju
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Chul Won Lee
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea.,MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Sang Chan Kim
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Won G An
- Institute of Marine Biotechnology, Pusan National University, Busan 609-735, Republic of Korea.,Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| |
Collapse
|