1
|
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: 2.3] [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
|
2
|
Bourdy G, Aubertin C, Jullian V, Deharo E. Quassia "biopiracy" case and the Nagoya Protocol: A researcher's perspective. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:290-297. [PMID: 28576580 DOI: 10.1016/j.jep.2017.05.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/26/2017] [Accepted: 05/27/2017] [Indexed: 06/07/2023]
Abstract
Biopiracy accusations are common in the world of biodiversity research. At the end of 2015, a French NGO accused researchers from the Institut de Recherche pour le Développement (IRD) of biopiracy. These researchers had applied for a patent for a natural bioactive molecule against malaria and cancer, the Simalikalactone E, isolated from Quassia amara L. (Simaroubaceae) leaves. This biopiracy allegation triggered a huge wave of attacks from the media and social networks, and vehement recrimination from political officials in French Guiana against researchers who have been accused of ethical misconduct, by stealing the traditional knowledge of indigenous people. These accusations were made in the contentious context of the ratification of the Nagoya Protocol in the frame of implementing the French law on biodiversity, nature and landscapes. So, in an atmosphere of heightened emotions it is crucial to understand the issues behind these accusations. We describe herein the genesis of our discovery, present the detractors' arguments, and discuss the consequences of such biopiracy denunciations for scientific research. We also address our concerns about the gap between rhetoric and reality and the real impact of the Nagoya Protocol on biodiversity conservation.
Collapse
Affiliation(s)
| | - Catherine Aubertin
- UMR 208 Patrimoines locaux et gouvernance, Institut de recherche pour le développement/Muséum national d'Histoire naturelle, 57 rue Cuvier - CP 51 75231, Paris cedex 05, France
| | - Valérie Jullian
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, France
| | - Eric Deharo
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, France..
| |
Collapse
|
3
|
Delmas A, Cherier J, Pohorecka M, Medale-Giamarchi C, Meyer N, Casanova A, Sordet O, Lamant L, Savina A, Pradines A, Favre G. The c-Jun/RHOB/AKT pathway confers resistance of BRAF-mutant melanoma cells to MAPK inhibitors. Oncotarget 2016; 6:15250-64. [PMID: 26098773 PMCID: PMC4558149 DOI: 10.18632/oncotarget.3888] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/25/2015] [Indexed: 11/25/2022] Open
Abstract
The response of BRAF-mutant melanoma patients to BRAF inhibitors is dramatically impaired by secondary resistances and rapid relapse. So far, the molecular mechanisms driving these resistances are not completely understood. Here, we show that, in BRAF-mutant melanoma cells, inhibition of BRAF or its target MEK induces RHOB expression by a mechanism that depends on the transcription factor c-Jun. In those cells, RHOB deficiency causes hypersensitivity to BRAF and MEK inhibitors-induced apoptosis. Supporting these results, loss of RHOB expression in metastatic melanoma tissues is associated with an increased progression-free survival of BRAF-mutant patients treated with vemurafenib. Following BRAF inhibition, RHOB activates AKT whose inhibition causes hypersensitivity of BRAF-mutant melanoma cells to BRAF inhibitors. In mice, AKT inhibition synergizes with vemurafenib to block tumor growth of BRAF-mutant metastatic melanoma. Our findings reveal that BRAF inhibition activates a c-Jun/RHOB/AKT pathway that promotes tumor cell survival and further support a role of this pathway in the resistance of melanoma to vemurafenib. Our data also highlight the importance of using RHOB tumor levels as a biomarker to predict vemurafenib patient's response and to select those that would benefit of the combination with AKT inhibitors.
Collapse
Affiliation(s)
- Audrey Delmas
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Julia Cherier
- Inserm, UMR 1037-CRCT, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Magdalena Pohorecka
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Claire Medale-Giamarchi
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Nicolas Meyer
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Department of Dermatology, Toulouse, France
| | - Anne Casanova
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Olivier Sordet
- Inserm, UMR 1037-CRCT, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Laurence Lamant
- Inserm, UMR 1037-CRCT, Toulouse, France.,Centre Hospitalo-Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Department of Pathology, Toulouse, France
| | - Ariel Savina
- Scientific Partnerships, Roche SAS, Boulogne Billancourt, France
| | - Anne Pradines
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| | - Gilles Favre
- Inserm, UMR 1037-CRCT, Toulouse, France.,Université Paul Sabatier, Toulouse, France.,Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Laboratory of Medical Biology and Oncogenetics, Toulouse, France
| |
Collapse
|
4
|
Le HL, Jullian V, Claparols C, Vansteelandt M, Haddad M, Cabou C, Deharo E, Fabre N. Development and validation of liquid chromatography combined with tandem mass spectrometry methods for the quantitation of simalikalactone E in extracts of Quassia amara L. and in mouse blood. PHYTOCHEMICAL ANALYSIS : PCA 2015; 26:111-118. [PMID: 25431121 DOI: 10.1002/pca.2542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 06/04/2023]
Abstract
INTRODUCTION Simalikalactone E (SkE) from Quassia amara, has been proved to be a valuable anti-malarial and anti-cancer compound. As SkE is very scarce, methods of quantitation are needed in order to optimise its isolation process and to determine pharmacokinetic data. OBJECTIVE To validate methods using liquid chromatography coupled to mass spectrometry for the quantitation of SkE in plant extracts and in biological fluids. METHODS High- and ultrahigh-performance liquid chromatography (UHPLC) coupled to ion trap mass spectrometry (MS) with single ion monitoring detection and to triple quadrupole-linear ion trap tandem mass spectrometry with multiple reaction monitoring detection methods were developed. Validation procedure was realised according to the International Conference on Harmonisation guideline. Methanol extracts of dried Quassia amara leaves, and mouse-blood samples obtained after various routes of administration, were analysed for SkE. RESULTS Methods were validated and gave similar results regarding the content of SkE expressed per kilogram of dry leaves in the traditional decoction (160 ± 12 mg/kg) and in the methanol extract (93 ± 2 mg/kg). The recovery of the analyte from mouse blood ranged from 80.7 to 119.8%. Simalikalactone E was only detected using UHPLC-MS/MS (0.2 ± 0.03 mg/L) in mouse blood after intravenous injection: none was detected following intraperitoneal or oral gavage administration of SkE. CONCLUSION The LC-MS methods were used for the quantitation of SkE in plant extracts and in mouse blood. These methods open the way for further protocol optimisation of SkE extraction and the determination of its pharmacokinetic data.
Collapse
Affiliation(s)
- Hong Luyen Le
- Université de Toulouse, UPS, UMR 152 Pharma-DEV, Université Toulouse 3, Faculté des Sciences Pharmaceutiques, F-31062, Toulouse cedex 09, France; Institut de Recherche pour le Développement (IRD), UMR 152 Pharma-DEV, F-31062, Toulouse cedex 09, France
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Pagani IS, Spinelli O, Mattarucchi E, Pirrone C, Pigni D, Amelotti E, Lilliu S, Boroni C, Intermesoli T, Giussani U, Caimi L, Bolda F, Baffelli R, Candi E, Pasquali F, Lo Curto F, Lanfranchi A, Porta F, Rambaldi A, Porta G. Genomic quantitative real-time PCR proves residual disease positivity in more than 30% samples with negative mRNA-based qRT-PCR in Chronic Myeloid Leukemia. Oncoscience 2014; 1:510-21. [PMID: 25594053 PMCID: PMC4278316 DOI: 10.18632/oncoscience.65] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 07/23/2014] [Indexed: 12/20/2022] Open
Abstract
Imatinib mesylate (IM) is the first line therapy against Chronic Myeloid Leukemia, effectively prolonging overall survival. Because discontinuation of treatment is associated with relapse, IM is required indefinitely to maintain operational cure. To assess minimal residual disease, cytogenetic analysis is insensitive in a high background of normal lymphocytes. The qRT-PCR provides highly sensitive detection of BCR-ABL1 transcripts, but mRNA levels are not directly related to the number of leukemic cells, and undetectable results are difficult to interpret. We developed a sensitive approach to detect the number of leukemic cells by a genomic DNA (gDNA) Q-PCR assay based on the break-point sequence, with a formula to calculate the number of Ph-positive cells. We monitored 8 CML patients treated with IM for more than 8 years. We tested each samples by patient specific gDNA Q-PCR in parallel by the conventional techniques. In all samples positive for chimeric transcripts we showed corresponding chimeric gDNA by Q-PCR, and in 32.8% (42/128) of samples with undetectable levels of mRNA we detected the persistence of leukemic cells. The gDNA Q-PCR assay could be a new diagnostic tool used in parallel to conventional techniques to support the clinician's decision to vary or to STOP IM therapy.
Collapse
Affiliation(s)
- Ilaria S Pagani
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy ; Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Orietta Spinelli
- Hematology laboratory, USC Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Elia Mattarucchi
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Cristina Pirrone
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Diana Pigni
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Elisabetta Amelotti
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Silvia Lilliu
- Hematology laboratory, USC Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Chiara Boroni
- Hematology laboratory, USC Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Tamara Intermesoli
- Hematology laboratory, USC Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Ursula Giussani
- Laboratory of Medical Genetics, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Luigi Caimi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Bolda
- Laboratory of chemical-clinical analysis, Section of Hematology and blood coagulation, Stem Cells laboratory, Spedali Civili of Brescia, Brescia, Italy
| | - Renata Baffelli
- Laboratory of chemical-clinical analysis, Section of Hematology and blood coagulation, Stem Cells laboratory, Spedali Civili of Brescia, Brescia, Italy
| | - Eleonora Candi
- Department of Experimental Medicine and Surgery, Tor Vergata University, Rome, Italy
| | - Francesco Pasquali
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Francesco Lo Curto
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| | - Arnalda Lanfranchi
- Laboratory of chemical-clinical analysis, Section of Hematology and blood coagulation, Stem Cells laboratory, Spedali Civili of Brescia, Brescia, Italy
| | - Fulvio Porta
- Laboratory of chemical-clinical analysis, Section of Hematology and blood coagulation, Stem Cells laboratory, Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Rambaldi
- Hematology laboratory, USC Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Giovanni Porta
- Department of Experimental and Clinical Medicine, Insubria University, Varese, Italy
| |
Collapse
|
6
|
Haagenson KK, Zhang JW, Xu Z, Shekhar MP, Wu GS. Functional analysis of MKP-1 and MKP-2 in breast cancer tamoxifen sensitivity. Oncotarget 2014; 5:1101-10. [PMID: 24658355 PMCID: PMC4011587 DOI: 10.18632/oncotarget.1795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 11/03/2014] [Indexed: 02/07/2023] Open
Abstract
Increased activation of ERK signaling has been reported in breast cancer models of acquired tamoxifen resistance. Here, we examined the expression of Mitogen-Activated Protein Kinase Phosphatases (MKPs) 1 and 2 following tamoxifen treatment and the effects of MKP-1/MKP-2 overexpression on tamoxifen sensitivity. Treatment of MCF7 breast cancer cells with tamoxifen increased MKP-2, but not MKP-1, protein levels. Overexpression of MKP-1 or MKP-2 inhibited estrogen-induced MCF7 cell proliferation compared to vector controls. MCF7-MKP-2 cells displayed significantly increased sensitivity to tamoxifen as compared to vector control or MCF7-MKP-1 cells. MKP-1 or MKP-2 overexpression eliminated ERK1/2 phosphorylation, suggesting that decreases in estrogen-induced proliferation of MKP-1 and MKP-2 overexpressing cells are due to ERK1/2 dephosphorylation. JNK1/2 activation was not detectable in any of these cells. These data suggest that tamoxifen-induced death of these cells is not dependent upon JNK signaling, but rather that ERK is the major MAPK driving their proliferation. MCF7-TAMR cells express higher levels of MKP-2 mRNA and protein than MCF7 cells. MKP-2 and phospho-ERK1/2 proteins are constitutively expressed in MCF7-TAMR cells, and activated JNK1/2 is not detectable. These data suggest that MKP-2 rather than MKP-1 is tamoxifen-regulated and that the elevated expression of MKP-2 in MCF7-TAMR cells potentially functions to restore tamoxifen sensitivity.
Collapse
Affiliation(s)
- Kelly K. Haagenson
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | | | - Zhengfan Xu
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Malathy P.V. Shekhar
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - Gen Sheng Wu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI
- Barbara Ann Karmanos Cancer Institute, Detroit, MI
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
7
|
Hairy cell leukemia: short review, today's recommendations and outlook. Blood Cancer J 2014; 4:e184. [PMID: 24531447 PMCID: PMC3944661 DOI: 10.1038/bcj.2014.3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/23/2013] [Indexed: 02/08/2023] Open
Abstract
Hairy cell leukemia (HCL) is part of the low-grade non-Hodgkin lymphoma family and represents approximately 2% of all leukemias. Treatment with splenectomy and interferon-α historically belonged to the first steps of therapeutic options, achieving partial responses/remissions (PR) in most cases with a median survival between 4 and 6 years in the 1980s. The introduction of the purine analogs (PA) pentostatin and cladribine made HCL a well-treatable disease: overall complete response rates (CRR) range from 76 to 98%, with a median disease-free survival (DFS) of 16 years a normal lifespan can be reached and HCL-related deaths are rare. However, insufficient response to PA with poorer prognosis and relapse rates of 30–40% after 5–10 years of follow-up may require alternative strategies. Minimal residual disease can be detected by additional examinations of bone marrow specimens after treatment with PA. The use of immunotherapeutic monoclonal antibodies (mAB) like rituximab as a single agent or in combination with a PA or more recently clinical trials with recombinant immunotoxins (RIT) show promising results to restrict these problems. Recently, the identification of the possible disease-defining BRAF V600E mutation may allow the development of new therapeutic targets.
Collapse
|
8
|
Xue P, Zhao Y, Liu Y, Yuan Q, Xiong C, Ruan J. A novel compound RY10-4 induces apoptosis and inhibits invasion via inhibiting STAT3 through ERK-, p38-dependent pathways in human lung adenocarcinoma A549 cells. Chem Biol Interact 2013; 209:25-34. [PMID: 24300195 DOI: 10.1016/j.cbi.2013.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/07/2013] [Accepted: 11/25/2013] [Indexed: 12/18/2022]
Abstract
Previous reports suggested that protoapigenone showed remarkable antitumor activities against a broad spectrum of human cancer cell lines, but had no effect on human lung adenocarcinoma A549 cell. The lack of effective remedies had necessitated the application of new therapeutic scheme. A novel compound RY10-4 which has the similar structure close to protoapigenone showed better antitumor activity. Treatment with RY10-4 inhibited the expression of pro-caspase-3, pro-caspase-9, Bcl-2 as well as phosphorylation of signal transducer and activator of transcription-3 (p-STAT3). It also reduced the expressions of matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9) and increases the expressions of reversion-inducing cysteine-rich protein with kazal motifs (RECK), as well as tissue inhibitor of metalloproteinase (TIMP) via inhibiting STAT3 by activating the mitogen-activated protein (MAP) kinases (the c-Jun N-terminal kinase (JNK), the p38 and extracellular signal-regulated kinase (ERK)) in A549 cells treated with RY10-4. Moreover, the cytotoxic effect of RY10-4 was induction of apoptosis in A549 cells by enhancing production of reactive oxygen species (ROS). Taken together, the observations suggested that RY10-4 had affected Bcl-2 family members, caspases, MMPs, TIMPs expressions and ROS production via inhibiting STAT3 activities through ERK and p38 pathways in A549 cells.
Collapse
Affiliation(s)
- Pingping Xue
- Key Laboratory of Natural Medicinal Chemistry and Resources Evaluation of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13# Hangkong Road, Wuhan 430030, PR China
| | - Yang Zhao
- Key Laboratory of Natural Medicinal Chemistry and Resources Evaluation of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13# Hangkong Road, Wuhan 430030, PR China
| | - Yang Liu
- School of Life Science, Wuchang University of Technology, Wuhan 430223, PR China
| | - Qianying Yuan
- Department of Pharmacology, Yale Medical School, New Haven, CT 06510, USA
| | - Chaomei Xiong
- Key Laboratory of Natural Medicinal Chemistry and Resources Evaluation of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13# Hangkong Road, Wuhan 430030, PR China
| | - Jinlan Ruan
- Key Laboratory of Natural Medicinal Chemistry and Resources Evaluation of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13# Hangkong Road, Wuhan 430030, PR China.
| |
Collapse
|
9
|
Abstract
Recent groundbreaking discoveries have revealed that IGF-1, Ras, MEK, AMPK, TSC1/2, FOXO, PI3K, mTOR, S6K, and NFκB are involved in the aging process. This is remarkable because the same signaling molecules, oncoproteins and tumor suppressors, are well-known targets for cancer therapy. Furthermore, anti-cancer drugs aimed at some of these targets have been already developed. This arsenal could be potentially employed for anti-aging interventions (given that similar signaling molecules are involved in both cancer and aging). In cancer, intrinsic and acquired resistance, tumor heterogeneity, adaptation, and genetic instability of cancer cells all hinder cancer-directed therapy. But for anti-aging applications, these hurdles are irrelevant. For example, since anti-aging interventions should be aimed at normal postmitotic cells, no selection for resistance is expected. At low doses, certain agents may decelerate aging and age-related diseases. Importantly, deceleration of aging can in turn postpone cancer, which is an age-related disease.
Collapse
|
10
|
Promotion of autophagy at the maturation step by IL-6 is associated with the sustained mitogen-activated protein kinase/extracellular signal-regulated kinase activity. Mol Cell Biochem 2013; 380:219-27. [PMID: 23677697 DOI: 10.1007/s11010-013-1676-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 05/02/2013] [Indexed: 12/19/2022]
Abstract
Increased autophagic vacuoles (AVs) occur in injured or degenerating neurons, under both developmental and pathological situations. Although an induced autophagy has been shown in inflammation response to cell factors, the underlying mechanism(s) remain(s) unknown. Here, we show that both cell factor IL-6 and environmental toxin MPP(+) promote the formation of vacuolation in SHSY5Y cells. By electron and immunofluorescent microscopy analyses, we showed that these structures are acid autolysosomes, containing cellular debris, and labeled by LC3 or LAMP1, markers of autophagosomes or lysosomes, respectively. Combining MPP(+) and IL-6 do not further increase vacuolation of SHSY5Y cells, and the vacuolation is less than that in the MPP(+)-treated group. MPP(+)-induced vacuolation results from significant increase in autophagy formation and delay in autophagy degradation, in relation to a decline of the lysosomal activity of arylsulfatase A. At molecular level, we show that this defect in autolysosomal maturation is independent of mammalian target of rapamycin and p38 inhibitions. Most importantly, we provide the first evidence that activation of ERK pathway is sufficient to commit cell to autophagic vacuolation. The sustained activation is required for MPP(+) to disrupt the autophagic pathway. IL-6 also induces a temporary and significant activation of ERK, but not sustained activation, and change sustained activation in MPP(+)-treated group into temporary activation. Taken together, these findings strongly support that IL-6 promotes the maturation of autophagosomes into functional autolysosomes by regulating ERK.
Collapse
|