1
|
Abi Sleiman M, Younes M, Hajj R, Salameh T, Abi Rached S, Abi Younes R, Daoud L, Doumiati JL, Frem F, Ishak R, Medawar C, Naim HY, Rizk S. Urtica dioica: Anticancer Properties and Other Systemic Health Benefits from In Vitro to Clinical Trials. Int J Mol Sci 2024; 25:7501. [PMID: 39000608 PMCID: PMC11242153 DOI: 10.3390/ijms25137501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
While conventional medicine has advanced in recent years, there are still concerns about its potential adverse reactions. The ethnopharmacological knowledge established over many centuries and the existence of a variety of metabolites have made medicinal plants, such as the stinging nettle plant, an invaluable resource for treating a wide range of health conditions, considering its minimal adverse effects on human health. The aim of this review is to highlight the therapeutic benefits and biological activities of the edible Urtica dioica (UD) plant with an emphasis on its selective chemo-preventive properties against various types of cancer, whereby we decipher the mechanism of action of UD on various cancers including prostate, breast, leukemia, and colon in addition to evaluating its antidiabetic, microbial, and inflammatory properties. We further highlight the systemic protective effects of UD on the liver, reproductive, excretory, cardiovascular, nervous, and digestive systems. We present a critical assessment of the results obtained from in vitro and in vivo studies as well as clinical trials to highlight the gaps that require further exploration for future prospective studies.
Collapse
Affiliation(s)
- Marc Abi Sleiman
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Maria Younes
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Roy Hajj
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Tommy Salameh
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Samir Abi Rached
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Rimane Abi Younes
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Lynn Daoud
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Jean Louis Doumiati
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Francesca Frem
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Ramza Ishak
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Christopher Medawar
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Hassan Y Naim
- Department of Biochemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Sandra Rizk
- Department of Natural Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| |
Collapse
|
2
|
DFF40 deficiency in cancerous T cells is implicated in chemotherapy drug sensitivity and resistance through the regulation of the apoptotic pathway. Biochem Pharmacol 2021; 194:114801. [PMID: 34678222 DOI: 10.1016/j.bcp.2021.114801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
The regulation of the apoptotic pathway is one of the most studied mechanisms regarding cancer cell resistance. Many mutations have been linked to drug resistance. The DNA fragmentation factor 40 (DFF40) has been gaining interest regarding cancer cell response to chemotherapy and patient outcomes. Glioblastomas and uterine leiomyosarcomas have been shown to have a downregulation of DFF40 expression, conferring a poor patient prognosis. In concordance with these observations, in this study, we showed that DFF40 gene is also downregulated in breast, endocervical, ovarian, lung, pancreas and glioblastomas. DFF40 is the endonuclease responsible of DNA fragmentation during apoptosis. In this study, we sought to determine if a DFF40 deficiency in Jurkat T cells could impact the sensitivity to conventional chemotherapy drugs. CRISPR-cas9 generated DFF40 knockout (DFF40 KO) stable Jurkat cells and wild-type (DFF40 WT) cells were treated with different antimetabolites and topoisomerase II (TOP2) inhibitors, and cell viability was subsequently assessed. DFF40 deficient cells show chemoresistance to antimetabolites (e.g. methotrexate, 6-mercaptopurine and cytarabine) and surprisingly, they are more sensitive to TOP2 inhibitors (e.g. etoposide and teniposide). DFF40 deficient cells exposed to cytarabine present lower phosphatidylserine translocation levels to the outer cell membrane layer. Etoposide exposure in DFF40 deficient cells induces higher mortality levels and downregulation of Bcl-xL cells compared to DFF40 expressing T cells. The abolition of DFF40 expression in Jurkat cells significantly impairs histone H2AX phosphorylation following etoposide and cytarabine treatments. Our findings suggest that DFF40 is a novel key target in cancer cell resistance that potentially regulates genomic stability.
Collapse
|
3
|
Kulbay M, Bernier-Parker N, Bernier J. The role of the DFF40/CAD endonuclease in genomic stability. Apoptosis 2021; 26:9-23. [PMID: 33387146 DOI: 10.1007/s10495-020-01649-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
Abstract
Maintenance of genomic stability in cells is primordial for cellular integrity and protection against tumor progression. Many factors such as ultraviolet light, oxidative stress, exposure to chemical reagents, particularly mutagens and radiation, can alter the integrity of the genome. Thus, human cells are equipped with many mechanisms that prevent these irreversible lesions in the genome, as DNA repair pathways, cell cycle checkpoints, and telomeric function. These mechanisms activate cellular apoptosis to maintain DNA stability. Emerging studies have proposed a new protein in the maintenance of genomic stability: the DNA fragmentation factor (DFF). The DFF40 is an endonuclease responsible of the oligonucleosomal fragmentation of the DNA during apoptosis. The lack of DFF in renal carcinoma cells induces apoptosis without oligonucleosomal fragmentation, which poses a threat to genetic information transfer between cancerous and healthy cells. In this review, we expose the link between the DFF and genomic instability as the source of disease development.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
- Department of Medicine, Université de Montréal, 2900 Blvd. Edouard Montpetit, Montreal, QC, Canada
| | - Nathan Bernier-Parker
- Toronto Animal Health Partners Emergency and Specialty Hospital, 1 Scarsdale Road, North York, ON, M3B 2R2, Canada
| | - Jacques Bernier
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
4
|
Kulbay M, Johnson B, Bernier J. DNA fragmentation factor 40 expression in T cells confers sensibility to tributyltin-induced apoptosis. Toxicology 2019; 426:152255. [PMID: 31401084 DOI: 10.1016/j.tox.2019.152255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
Abstract
DNA fragmentation factor 40 (DFF40), an endonuclease, mediates the final and irreversible step of apoptosis by conducting oligonucleosomal DNA fragmentation. New emerging studies have proposed a role of DFF40 in genomic stability, besides its nuclease activity. Overexpression of DFF40 in tumoral cells increases their sensitivity to chemotherapeutic drugs. In this study, we sought to determine if DFF40 expression influences the toxicity of tributyltin (TBT), a well-known immunotoxic and apoptosis-inducing compound. The strategy used was to knockout DFF40 expression by CRISPR-cas9 method in Jurkat T cells and to determine the toxicity of TBT in DFF40 KO cells and DFF40 WT Jurkat cells. DFF40 KO Jurkat cells show an increase of cell viability following a 24-h TBT exposure (p < 0.05). There is a resistance to TBT-induced apoptosis determined by annexin V/PI am labeling (p < 0.05). Interestingly, the basal level of ROS rises in DFF40 KO Jurkat cells, but ROS production levels after TBT exposure remains at the same basal level. Other apoptosis or DNA damage makers (procaspase-3, caspase-6, and PARP cleavage) are significantly delayed and decreased. DFF40 deficient cells do not present histone H2AX phosphorylation, whereas wild-type cells present a phosphorylation following a 6-h exposure to TBT (p < 0.001). The re-expression of DFF40 in DFF40 KO cells restores the cytotoxic effects of TBT. Overall, these data suggest a role of DFF40 in cells sensitivity to TBT and possibly in DNA stability.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, H7V 1B7, Laval, Québec, Canada
| | - Bruno Johnson
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, H7V 1B7, Laval, Québec, Canada
| | - Jacques Bernier
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, H7V 1B7, Laval, Québec, Canada.
| |
Collapse
|
5
|
Banas T, Pitynski K, Okon K, Winiarska A. Non-endometrioid and high-grade endometrioid endometrial cancers show DNA fragmentation factor 40 (DFF40) and B-cell lymphoma 2 protein (BCL2) underexpression, which predicts disease-free and overall survival, but not DNA fragmentation factor 45 (DFF45) underexpression. BMC Cancer 2018; 18:418. [PMID: 29653556 PMCID: PMC5899339 DOI: 10.1186/s12885-018-4333-6] [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: 06/12/2017] [Accepted: 04/04/2018] [Indexed: 12/23/2022] Open
Abstract
Background The expression of DNA fragmentation factor 45 (DFF45) and B-cell lymphoma 2 (BCL2) in glands of the normal human endometrium is related to phases of the menstrual cycle and decreases after menopause, whereas the expression of DNA fragmentation factor 40 (DFF40) is stable. Moreover, DF45, BCL2 and DFF40 underexpression has been reported in numerous malignancies, including uterine leiomyosarcomas. In this study, we aimed to investigate DFF45, BCL2 and DFF40 expression in endometrioid and non-endometrioid types of endometrial cancers (ECs). We also evaluated the correlations between DFF45, BCL2 and DFF40 expression levels and clinicopathological parameters and determined the value of these three proteins as prognostic markers of disease-free survival (DFS) and overall survival (OS). Methods Immunohistochemistry was performed to evaluate DFF45, BCL2 and DFF40 expression in 342 cases of ECs. Student’s t-test, the Mann-Whitney U-test, and the chi-squared test were used for the statistical analyses as appropriate. The Cox-Mantel test, Cox’s proportional hazard model, and relative risk analyses were used to evaluate associations between DFF40, DFF45, and BCL2 expression and clinicopathological characteristics. Results DFF40 and BCL2, but not DFF45, were significantly underexpressed in non-endometrioid and high-grade endometrioid ECs compared with low- and moderate-grade endometrioid ECs. Women with DFF40- and BCL2-negative tumors had higher risks of disease recurrence, lymph node involvement, lympho-vascular space infiltration, and deep myometrial invasion compared with women with DFF40- and BCL2-positive tumors. Additionally, women with DFF40- and BCL2-negative tumors had significantly lower OS and DFS than women with DFF40- and BCL2-positive tumors. A multivariable analysis of the model, including the clinicopathological characteristics and immunohistochemical results, showed that negative BCL2 expression, lymph node involvement, and high-stage and high-grade disease were independent predictors of OS, whereas negative BCL2 expression, lymph node involvement, and high-stage disease were independent predictors of DFS. Conclusions Compared with low- and moderate-grade endometrioid ECs, non-endometrioid and high-grade endometrioid ECs showed significant DFF40 and BCL2 underexpression. The absence of DFF40 and BCL2 expression negatively affects DFS and OS. Further prospective studies are warranted to assess the potential utility of DFF40 and BCL2 as targets in the diagnosis or treatment of ECs.
Collapse
Affiliation(s)
- Tomasz Banas
- Department of Gynecology and Oncology, Jagiellonian University, 21 Kopernika Street, 30-501, Krakow, Poland.
| | - Kazimierz Pitynski
- Department of Gynecology and Oncology, Jagiellonian University, 21 Kopernika Street, 30-501, Krakow, Poland
| | - Krzysztof Okon
- Department of Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| | - Aleksandra Winiarska
- Department of Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
6
|
Transcriptome analysis of ABCB1, ABCG2 and the BCL2/BAX ratio in refractory and relapsed canine lymphomas under treatment and rescue protocol. ACTA VET-BEOGRAD 2018. [DOI: 10.2478/acve-2018-0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abstract
The main problems that cause unresponsiveness to an anti-neoplastic drug are the overexpression of drug resistant and anti-apoptotic proteins in tumor cells. In a rescue protocol we evaluated the ability of toceranib phosphate concurrent with lomustine (CCNU) or L-asparaginase and vincristine to decrease drug resistant and apoptotic proteins in relapsed and refractory canine lymphomas. The peripheral blood samples were collected before and after the rescue treatment from fourteen dogs that were refractory to cyclophosphamide-vincristine-prednisolone (COP) or COP-doxorubicin (CHOP) treatment and had recurrent multicentric lymphoma. The mRNA expression level of ABCB1, ABCG2, Bcl2 and Bax were determined by quantitative real-time PCR. The fold-change in ABCB1, ABCG2, Bcl2 and Bax mRNA levels were analyzed in correlation with the progression-free survival (PFS). After the rescue treatment, the ABCB1 and ABCG2 mRNA expression levels were 1.57- and 1.85-fold lower (p = 0.4 and p = 0.87), respectively, compared to pre-treatment. Bcl2/Bax ratio was numerically but not significantly decreased 1.02-fold (p = 0.74). The overall response rate of this protocol was 50% with a median PFS of 79 days (range 14-207 days). The low medians of relative expression levels of ABCB1, ABCG2 and Bcl2/Bax ratio group did not correlate with the clinical outcomes when compared to the high medians of relative expression levels, and likewise with the clinical stage, immunophenotype, histological grade and sub-stage. Therefore, the administration of a rescue drug with toceranib phosphate might be beneficial in refractory and relapsed canine lymphoma.
Collapse
|
7
|
Thomadaki E, Havredaki M, Tsiapalis CM. PAP Modulations in Daudi Cells and Molt-3 Cells Treated with Etoposide are Mutually Associated with Morphological Evidence of Apoptosis. Int J Biol Markers 2018; 19:203-12. [PMID: 15503822 DOI: 10.1177/172460080401900305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Daudi (B-cell line) and Molt-3 (T-cell line) cells provide a model for the study of apoptosis, the induction of which is often accompanied by concominant modulations of proteins involved in mRNA maturation. One of these proteins is poly(A) polymerase (PAP), which is responsible for mRNA cleavage and polyadenylation. A number of recent reports also suggest involvement of mRNA maturation and stability in the induction of specific pathways of cell apoptosis. In this study we identified PAP activity levels and isoform modulations in two different cell lines (Daudi and Molt-3) and related them to DNA fragmentation (a hallmark of apoptosis) and cell cycle phase specificity in terms of the temporal sequence of events and the time that elapsed between administration of the apoptosis inducer (the widely used anticancer drug etoposide) and the observed effects. Treatment of both cell lines with 20 μg/mL etoposide induced apoptosis after four hours in Molt-3 cells and only after 24 hours in Daudi cells, as revealed by two independent methods. In Daudi cells the PAP activity levels and isoforms were downregulated prior to ΔΨm reduction, DNA fragmentation and the morphological changes of the nucleus, whereas in Molt-3 cells no PAP activity and isoform modulations were observed prior to the early hallmarks of apoptosis.
Collapse
Affiliation(s)
- E Thomadaki
- Institute of Biology, NCSR Demokritos, Aghia Paraskevi Attikis, Athens--Greece
| | | | | |
Collapse
|
8
|
Targeting the metabolic pathway of human colon cancer overcomes resistance to TRAIL-induced apoptosis. Cell Death Discov 2016; 2:16067. [PMID: 27648301 PMCID: PMC5018545 DOI: 10.1038/cddiscovery.2016.67] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 01/05/2023] Open
Abstract
Colon cancer is a leading cause of cancer-related mortality for which targeted therapy is needed; however, trials using apoptosis-inducing ligand monotherapy to overcome resistance to apoptosis have not shown clinical responses. Since colon cancer cells selectively uptake and rapidly metabolize glucose, a property utilized for clinical staging, we investigated mechanisms to alter glucose metabolism in order to selectively target the cancer cells and to overcome evasion of apoptosis. We demonstrate TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) resistance in the majority of human colon cancers tested and utilize the glucose analog 2-deoxy-d-glucose to sensitize TRAIL-resistant gastrointestinal adenocarcinoma cells, and not normal gastrointestinal epithelial cells, to TRAIL-induced apoptosis through enhanced death receptor 5 expression, downstream modulation of MAPK signaling and subsequent miRNA expression modulation by increasing the expression of miR-494 via MEK activation. Further, established human colon cancer xenografts treated with this strategy experience anti-tumor responses. These findings in colon adenocarcinoma support further investigation of manipulation of cellular energetics to selectively overcome resistance to apoptosis and to impart tumor regressions in established colon cancer tumors.
Collapse
|
9
|
Radiotherapy and TRAIL for cancer therapy. Cancer Lett 2011; 332:184-93. [PMID: 21824725 DOI: 10.1016/j.canlet.2011.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 06/10/2011] [Accepted: 07/02/2011] [Indexed: 11/22/2022]
Abstract
The use of radiotherapy and concomitant chemotherapy substantially improved cure rates in patients with different malignant tumours. However, it is unlikely that further improvements based on conventional chemotherapy may be achieved in the future since increased rates of acute side effects already limit the value of these approaches. Additionally, the increased local control rates are counterweighted by still high rates of distant failures resulting in low net gains for the patients. Thus, there is a currently unmet need for the integration of target-specific drugs improving local control as well distant control into radiation based treatment protocols. In this regard, the death-receptor ligand TNF-α-related apoptosis-inducing ligand (TRAIL/Apo2L) and TRAIL-receptor agonistic antibodies were shown to display a high selectivity for tumour cells and act synergistically with conventional chemotherapy drugs and radiation. Up to now it has been shown that radiation strongly sensitises malignant cells to TRAIL and TRAIL-agonistic antibodies. Synergistic induction of apoptosis was demonstrated in a majority of malignant cell types and xenograft models. Especially in those cells types displaying only weak responses to either treatment alone, strong sensitising effects were described. Moreover, in merely all normal cells and tissues no synergistic effects were found. Depending on cell type and experimental setting, the efficacy of combined treatment is determined by the p53-status, the balance between pro- and anti-apoptotic Bcl-2 proteins and modulation of TRAIL-receptor signal transduction.
Collapse
|
10
|
Potent antitumoral activity of TRAIL through generation of tumor-targeted single-chain fusion proteins. Cell Death Dis 2010; 1:e68. [PMID: 21364672 PMCID: PMC3032523 DOI: 10.1038/cddis.2010.45] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In an attempt to improve TRAIL's (tumor necrosis factor-related apoptosis-inducing ligand) tumor selective activity a variant was designed, in which the three TRAIL protomers are expressed as a single polypeptide chain (scTRAIL). By genetic fusion with a single-chain antibody fragment (scFv) recognizing the extracellular domain of ErbB2, we further equipped scTRAIL with tumor-targeting properties. We studied tumor targeting and apoptosis induction of scFv–scTRAIL in comparison with non-targeted scTRAIL. Importantly, the tumor antigen-targeted scTRAIL fusion protein showed higher apoptotic activity in vitro, with a predominant action by TRAIL-R2 signaling. Pharmacokinetic studies revealed increased plasma half-life of the targeted scTRAIL fusion protein compared with scTRAIL. In vivo studies in a mouse tumor model with xenotransplanted Colo205 cells confirmed greater response to the ErbB2-specific scTRAIL fusion protein compared with non-targeted scTRAIL both under local and systemic application regimen. Together, in vitro and in vivo data give proof of concept of higher therapeutic activity of tumor-targeted scFv–scTRAIL molecules. Further, we envisage that through targeting of scTRAIL, potential side effects should be minimized. We propose that scFv-mediated tumor targeting of single-chain TRAIL represents a promising strategy to improve TRAIL's antitumoral action and to minimize potential unwanted actions on normal tissues.
Collapse
|
11
|
Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene 2009; 29:1641-52. [PMID: 19966861 DOI: 10.1038/onc.2009.448] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Most cancer cells exhibit increased glycolysis for generation of their energy supply. This specificity could be used to preferentially kill these cells. In this study, we identified the signaling pathway initiated by glycolysis inhibition that results in sensitization to death receptor (DR)-induced apoptosis. We showed, in several human cancer cell lines (such as Jurkat, HeLa, U937), that glucose removal or the use of nonmetabolizable form of glucose (2-deoxyglucose) dramatically enhances apoptosis induced by Fas or by tumor necrosis factor-related apoptosis-inducing ligand. This sensitization is controlled through the adenosine monophosphate (AMP)-activated protein kinase (AMPK), which is the central energy-sensing system of the cell. We established the fact that AMPK is activated upon glycolysis block resulting in mammalian target of rapamycin (mTOR) inhibition leading to Mcl-1 decrease, but no other Bcl-2 anti-apoptotic members. Interestingly, we determined that, upon glycolysis inhibition, the AMPK-mTOR pathway controlled Mcl-1 levels neither through transcriptional nor through posttranslational mechanism but rather by controlling its translation. Therefore, our results show a novel mechanism for the sensitization to DR-induced apoptosis linking glucose metabolism to Mcl-1 downexpression. In addition, this study provides a rationale for the combined use of DR ligands with AMPK activators or mTOR inhibitors in the treatment of human cancers.
Collapse
|
12
|
Marini P, Junginger D, Stickl S, Budach W, Niyazi M, Belka C. Combined treatment with lexatumumab and irradiation leads to strongly increased long term tumour control under normoxic and hypoxic conditions. Radiat Oncol 2009; 4:49. [PMID: 19860913 PMCID: PMC2772841 DOI: 10.1186/1748-717x-4-49] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 10/27/2009] [Indexed: 01/15/2023] Open
Abstract
Purpose The combination of ionizing radiation with the pro-apoptotic TRAIL receptor antibody lexatumumab has been shown to exert considerable synergistic apoptotic effects in vitro and in short term growth delay assays. To clarify the relevance of these effects on local tumour control long-term experiments using a colorectal xenograft model were conducted. Materials and methods Colo205-xenograft bearing NMRI (nu/nu) nude mice were treated with fractionated irradiation (5× 3 Gy, d1-5) and lexatumumab (0.75 mg/kg, d1, 4 and 8). The tumour bearing hind limbs were irradiated with graded single top up doses at d8 under normoxic (ambient) and acute hypoxic (clamped) conditions. Experimental animals were observed for 270 days. Growth delay and local tumour control were end points of the study. Statistical analysis of the experiments included evaluation of tumour regrowth and local tumour control. Results Combined treatment with irradiation and lexatumumab led to a pronounced tumour regrowth-delay when compared to irradiation alone. The here presented long-term experiments revealed a highly significant rise of local tumour control for normoxic (ambient) (p = 0. 000006) and hypoxic treatment (p = 0. 000030). Conclusion Our data show that a combination of the pro-apoptotic antibody lexatumumab with irradiation reduces tumour regrowth and leads to a highly increased local tumour control in a nude mouse model. This substantial effect was observed under ambient and more pronounced under hypoxic conditions.
Collapse
Affiliation(s)
- Patrizia Marini
- CCC Tübingen, Dept of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str 3, 72076 Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
13
|
Combination of the pro-apoptotic TRAIL-receptor antibody mapatumumab with ionizing radiation strongly increases long-term tumor control under ambient and hypoxic conditions. Int J Radiat Oncol Biol Phys 2009; 75:198-202. [PMID: 19695436 DOI: 10.1016/j.ijrobp.2009.04.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 04/16/2009] [Accepted: 04/16/2009] [Indexed: 11/22/2022]
Abstract
PURPOSE Mapatumumab, an agonistic tumor necrosis factor-related apoptosis inducing ligand-receptor antibody, exerts highly synergistic apoptotic effects in vitro and in short-term growth delay assays when combined with irradiation. Because it remained unclear in how far these effects influence local tumor control, long-term experiments using a colorectal xenograft model were undertaken. MATERIAL AND METHODS Experiments were performed with irradiation (5 x 3 Gy, d1-5) and mapatumumab (10 mg/kg) in Colo205-xenograft-bearing NMRI (nu/nu) nude mice. Graded top up doses were delivered on the tumor-bearing hind leg under ambient and hypoxic conditions; follow-up was 270 days. Growth delay and local tumor control were end points of the study. Statistical analysis of the experiments included calculation of tumor regrowth and local tumor control. RESULTS After combined treatment, a pronounced tumor regrowth-delay was observed when compared with irradiation alone. Long-term experiments revealed a highly significant increase in local tumor control for ambient (p = 0.00076) and hypoxic treatment (p = 0.000069). CONCLUSIONS The present data demonstrate for the first time that combination of a pro-apoptotic antibody with irradiation results in evidently reduced tumor regrowth times and subsequently highly increased local tumor control under normoxic and hypoxic conditions in a xenograft mouse model.
Collapse
|
14
|
Essafi-Benkhadir K, Grosso S, Puissant A, Robert G, Essafi M, Deckert M, Chamorey E, Dassonville O, Milano G, Auberger P, Pagès G. Dual role of Sp3 transcription factor as an inducer of apoptosis and a marker of tumour aggressiveness. PLoS One 2009; 4:e4478. [PMID: 19212434 PMCID: PMC2636865 DOI: 10.1371/journal.pone.0004478] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 01/08/2009] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The ambiguous role of transcription factor Sp3 for tumour progression is still debated since it was described as a transcriptional repressor or activator. Here we tried to decipher the molecular mechanisms implicated in Sp3 accumulation observed in aggressive tumours. METHODOLOGY We generated normal and tumour cell lines conditionally expressing Sp3. Cell growth was analyzed in vitro and after inoculation in nude mice. Apoptosis was assessed by pan- caspase activity assays, by counting fragmented nuclei and by determination of caspase 9 cleavage. Gene expression was determined by quantitative PCR. Cleavage by different caspases was performed after in vitro translation of the Sp3 cDNA in the presence of [S(35)] labelled methionine. Different tumour cell lines and head and neck tumour samples were tested for the presence of Sp3 by western blots. Correlation between Sp3 expression and overall survival has been statistically determined. PRINCIPAL FINDINGS Conditional over-expression of Sp3 induces apoptosis and modifies expression of genes implicated in the regulation of cell cycle and pro and anti apoptotic genes. Sp3 over-expression strongly reduces the development of tumours in nude mice confirming its pro-apoptotic potential in vivo. However, cells can survive to apoptosis through selective Sp3 cleavage by caspase. Sp3 induction in established tumours resulted in transient regression then progression. Progression coincides with re-accumulation of the full length form of Sp3. Sp3 is over-expressed in tumour cell lines of different origins. The presence of high levels of the full-length form of Sp3 indicates a poor prognosis for overall survival of patients with head and neck tumours. CONCLUSIONS Full length Sp3 accumulation highlights bypass of tumour cell apoptotic capacities and is indicative of head and neck tumours aggressiveness.
Collapse
Affiliation(s)
- Khadija Essafi-Benkhadir
- University of Nice-Sophia Antipolis, Institute of Developmental Biology and Cancer Research UMR CNRS 6543, Centre Antoine Lacassagne, Nice, France
| | - Sébastien Grosso
- University of Nice-Sophia Antipolis, INSERM, U895, Cell Death Differentiation and Cancer Team, Equipe labellisée par la Ligue Nationale contre le Cancer, Faculty of Medicine, Nice, France
| | - Alexandre Puissant
- University of Nice-Sophia Antipolis, INSERM, U895, Cell Death Differentiation and Cancer Team, Equipe labellisée par la Ligue Nationale contre le Cancer, Faculty of Medicine, Nice, France
| | - Guillaume Robert
- University of Nice-Sophia Antipolis, INSERM, U895, Cell Death Differentiation and Cancer Team, Equipe labellisée par la Ligue Nationale contre le Cancer, Faculty of Medicine, Nice, France
| | - Makram Essafi
- University of Nice-Sophia Antipolis, INSERM, U576, Regulation of immunity and inflammatory reactions, Nice, France
| | - Marcel Deckert
- University of Nice-Sophia Antipolis, INSERM, U576, Regulation of immunity and inflammatory reactions, Nice, France
| | | | | | - Gérard Milano
- Oncopharmacology unit (EA 3836), Centre Antoine Lacassagne, Nice, France
| | - Patrick Auberger
- University of Nice-Sophia Antipolis, INSERM, U895, Cell Death Differentiation and Cancer Team, Equipe labellisée par la Ligue Nationale contre le Cancer, Faculty of Medicine, Nice, France
| | - Gilles Pagès
- University of Nice-Sophia Antipolis, Institute of Developmental Biology and Cancer Research UMR CNRS 6543, Centre Antoine Lacassagne, Nice, France
- * E-mail:
| |
Collapse
|
15
|
Desharnais P, Dupéré-Minier G, Hamelin C, Devine P, Bernier J. Involvement of CD45 in DNA fragmentation in apoptosis induced by mitochondrial perturbing agents. Apoptosis 2008; 13:197-212. [PMID: 18157742 DOI: 10.1007/s10495-007-0162-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CD45 is a type I transmembrane molecule with phosphatase activity which comprises up to 10% of the cell surface area in nucleated haematopoietic cells. We have previously demonstrated the absence of nuclear apoptosis in CD45-negative T cells after chemical-induced apoptosis. The aim of this study was to characterize the role of CD45 in nuclear apoptosis. In contrast to wild type CD45-positive T cells, the CD45-deficient T cell lines are resistant to the induction of DNA fragmentation and chromatin condensation following tributyltin (TBT) or H2O2 exposure, but not to cycloheximide-induced apoptosis. CD45 transfection in deficient cell lines led to the restoration of chromatin condensation and DNA fragmentation following TBT exposure. In both CD45-positive and negative T cell lines, TBT exposure mediates intracellular calcium mobilization, caspase-3 activation and DFF45 cleavage. Moreover, DNA fragmentation was also induced by TBT in cells deficient in expression of p56lck, ZAP-70 and SHP-1. Subcellular partitioning showed a decrease in nuclear localisation of caspase-3 and DFF40. Together, these results demonstrate for the first time, that CD45 expression plays a key role in internucleosomal DNA fragmentation and chromatin condensation processes during apoptosis. CD45 activity or its substrates' activity, appears to be located downstream of caspase-3 activation and plays a role in retention of DFF40 in the nucleus.
Collapse
Affiliation(s)
- Philippe Desharnais
- INRS-Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC, Canada, H7V 1B7
| | | | | | | | | |
Collapse
|
16
|
Barbarotto E, Corallini F, Rimondi E, Fadda R, Mischiati C, Grill V, Vaccarezza M, Celeghini C. Differential effects of chemotherapeutic drugs versus the MDM-2 antagonist nutlin-3 on cell cycle progression and induction of apoptosis in SKW6.4 lymphoblastoid B-cells. J Cell Biochem 2008; 104:595-605. [DOI: 10.1002/jcb.21649] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Cillessen SAGM, Meijer CJLM, Ossenkoppele GJ, Castricum KCM, Westra AH, Niesten P, Muris JJF, Nijdam HF, van der Hem KG, Flens M, Hooijberg E, Oudejans JJ. Human soluble TRAIL/Apo2L induces apoptosis in a subpopulation of chemotherapy refractory nodal diffuse large B-cell lymphomas, determined by a highly sensitive in vitro apoptosis assay. Br J Haematol 2006; 134:283-93. [PMID: 16848771 DOI: 10.1111/j.1365-2141.2006.06186.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Resistance to chemotherapy in therapy-refractory diffuse large B-cell lymphomas (DLBCL) is related to inhibition of the intrinsic apoptosis pathway. Human soluble tumour necrosis factor (TNF)-related apoptosis-inducing ligand (hsTRAIL/Apo2L) induces apoptosis via the alternative, death-receptor mediated apoptosis pathway and might be an effective alternative form of therapy for these lymphomas. This study investigated whether hsTRAIL/Apo2L could actually induce apoptosis in isolated lymphoma cells of DLBCL biopsies of patients with chemotherapy-refractory DLBCL. Twelve out of a total of 22 DLBCL samples were sensitive to hsTRAIL/Apo2L. These sensitive lymphomas included seven clinically chemotherapy-refractory lymphomas. Furthermore, hsTRAIL/Apo2L induced apoptosis in DLBCL cells and in B-cell lines that showed high expression levels of inhibitors of the intrinsic apoptosis pathway: Bcl-2 and/or X-linked inhibitor of apoptosis (XIAP). hsTRAIL/Apo2L-sensitive lymphoma cells showed expression of the TRAIL receptors R1 and/or R2 and absence of R3 and R4. We conclude that hsTRAIL/Apo2L induced apoptosis in a subpopulation of chemotherapy-refractory nodal DLBCL and that disruption of the intrinsic apoptosis-mediated pathway and expression of Bcl-2 and XIAP did not confer resistance to hsTRAIL/Apo2L-induced apoptosis in DLBCL. Thus, based on our results, further exploration of hsTRAIL/Apo2L as an alternative treatment for patients with chemotherapy-refractory DLBCL should be considered.
Collapse
Affiliation(s)
- Saskia A G M Cillessen
- Department of Clinical Pathology, VU Universrity Medical Centre, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Marini P, Denzinger S, Schiller D, Kauder S, Welz S, Humphreys R, Daniel PT, Jendrossek V, Budach W, Belka C. Combined treatment of colorectal tumours with agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and radiotherapy: enhanced effects in vitro and dose-dependent growth delay in vivo. Oncogene 2006; 25:5145-54. [PMID: 16636678 DOI: 10.1038/sj.onc.1209516] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We and others have demonstrated already that TRAIL (TNF-related apoptosis-inducing ligand) is a very promising candidate for molecular targeted anticancer therapy, especially when combined with ionizing radiation or other DNA-damaging agents. Agonist monoclonal antibodies that activate and are specific for the death signaling TRAIL receptors are an alternative method to stimulate the programmed cell death pathway. Phase 1 clinical trials have subsequently been conducted and shown a very good tolerability of these antibodies. In order to assess the efficacy of TRAIL receptor stimulation to induce cell death by this alternate method, we studied the combination of the agonistic-TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 with radiation in vitro and in vivo. Induction of apoptosis after combined treatment with TRAIL receptor antibodies HGS-ETR1 and/or HGS-ETR2 (0.01, 0.1, 1.0 mg/ml) and irradiation with 2, 5 or 10 Gy was determined by fluorescence microscopy and Western blot analysis of caspase-8 and PARP. The colorectal tumour cell lines Colo 205, HCT 116 and HCT-15 were used for in vitro experiments. Growth delay experiments were performed with combined treatment with fractionated irradiation (days 1-5 and 3 Gy single dose/day) and the receptor antibodies (intraperitonially, three different concentrations, application on days 1, 4 and 8) on Colo 205 xenograft-bearing NMRI (nu/nu) nude mice. HGS-ETR1 and HGS-ETR2 induced apoptotic cell death in a dose-dependent fashion and significantly increased cell death in combination with irradiation in vitro when compared to either irradiation or antibody treatment alone. The efficacy of the combined treatment seems to be at least partially Bax-dependent. Similar to the results from cell culture experiments, in vivo experiments demonstrated a dose-dependent delay in tumour growth after combined treatment. In vivo, in the Colo205 xenograft model, HGS-ETR2 revealed a higher activity than HGS-ETR1. This is the first study to demonstrate significant efficacy of combined treatment with the monoclonal agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and ionising radiation in in vitro and in vivo models. We postulate that HGS-ETR1 and HGS-ETR2 will be very promising new agents in the field of molecular targeted multi-modality anticancer therapy.
Collapse
Affiliation(s)
- P Marini
- CCC Tübingen, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bourcier C, Jacquel A, Hess J, Peyrottes I, Angel P, Hofman P, Auberger P, Pouysségur J, Pagès G. p44 Mitogen-Activated Protein Kinase (Extracellular Signal-Regulated Kinase 1)–Dependent Signaling Contributes to Epithelial Skin Carcinogenesis. Cancer Res 2006; 66:2700-7. [PMID: 16510590 DOI: 10.1158/0008-5472.can-05-3129] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular signal-regulated kinases (ERK) regulate cellular functions in response to a variety of external signals. However, the specific functions of individual ERK isoforms are largely unknown. Hence, we have investigated the specific function of ERK1 in skin homeostasis and tumorigenesis in ERK1 knockout mice. They spontaneously develop cutaneous lesions and hyperkeratosis with epidermis thickness. Skin hyperproliferation and inflammation induced by application of 12-O-tetradecanoylphorbol-13-acetate (TPA) is strongly reduced in mutant mice. ERK1(-/-) mice are resistant to development of skin papillomas induced by 7,12-dimethylbenz(a)anthracene (DMBA) and promoted by TPA. Tumor appearance was delayed, their formation was less frequent, and their number and size were reduced. Keratinocytes obtained from knockout mice showed reduced growth and resistance to apoptotic signals, accompanied by an impaired expression of genes implicated in growth control and invasiveness. These results highlight the importance of ERK1 in skin homeostasis and in the process of skin tumor development.
Collapse
Affiliation(s)
- Christine Bourcier
- Institute of Signaling, Developmental Biology and Cancer Research, UMR Centre National de la Recherche Scientifique, Nice, France
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Boulares H, Zoltoski A, Kandan S, Akbulut T, Yakovlev A, Oumouna M. Correlation between decreased sensitivity of the Daudi lymphoma cells to VP-16-induced apoptosis and deficiency in DNAS1L3 expression. Biochem Biophys Res Commun 2006; 341:653-62. [PMID: 16427601 DOI: 10.1016/j.bbrc.2006.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Accepted: 01/03/2006] [Indexed: 01/01/2023]
Abstract
A functional relationship between the apoptotic endonuclease DNAS1L3 and the chemotherapeutic drug VP-16 was established. The lymphoma cell line, Daudi, exhibited a significant resistance to VP-16 treatment in comparison to the lymphoma/leukemia cell line, U-937. While U-937 cells degraded their DNA into internucleosomal fragments, Daudi cells failed to undergo such fragmentation in response to the drug. Activation of both caspase-3 and DNA fragmentation factor was not sufficient to trigger internucleosomal DNA fragmentation in Daudi cells. No correlation was found between expression levels of topoisomerase-II, Pgp, Bcl-2, Bax, or Bad and decreased sensitivity of Daudi cells to VP-16. Daudi cells failed to express DNAS1L3 and ectopic expression of this protein significantly sensitized the cells to VP-16. An enhancement of caspase-3 activity and collapse of mitochondrial membrane potential underlie DNAS1L3-mediated sensitization of Daudi cells to VP-16, which may be a direct result of DNAS1L3-mediated increase in PARP-1-activating DNA breaks after VP-16 treatment. Our results suggest that DNAS1L3 plays an active role in lymphoma cell sensitization to VP-16 and that its deficiency may constitute a novel mechanism of drug resistance in these cells.
Collapse
Affiliation(s)
- Hamid Boulares
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Shiokawa D, Tanuma S. Differential DNases are selectively used in neuronal apoptosis depending on the differentiation state. Cell Death Differ 2004; 11:1112-20. [PMID: 15167901 DOI: 10.1038/sj.cdd.4401454] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In this study, we investigate the roles of two apoptotic endonucleases, CAD and DNase gamma, in neuronal apoptosis. High expression of CAD, but not DNase gamma, is detected in proliferating N1E-115 neuroblastoma cells, and apoptotic DNA fragmentation induced by staurosporine under proliferating conditions is abolished by the expression of a caspase-resistant form of ICAD. After the induction of neuronal differentiation, CAD disappearance and the induction of DNase gamma occur simultaneously in N1E-115 cells. Apoptotic DNA fragmentation that occurs under differentiating conditions is suppressed by the downregulation of DNase gamma caused by its antisense RNA. The induction of DNase gamma is also observed during neuronal differentiation of PC12 cells, and apoptotic DNA fragmentation induced by NGF deprivation is inhibited by the antisense-mediated downregulation of DNase gamma. These observations suggest that DNA fragmentation in neuronal apoptosis is catalyzed by either CAD or DNase gamma depending on the differentiation state. Furthermore, DNase gamma is suggested to be involved in naturally occurring apoptosis in developing nervous systems.
Collapse
Affiliation(s)
- D Shiokawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | |
Collapse
|
22
|
Luciano F, Jacquel A, Colosetti P, Herrant M, Cagnol S, Pages G, Auberger P. Phosphorylation of Bim-EL by Erk1/2 on serine 69 promotes its degradation via the proteasome pathway and regulates its proapoptotic function. Oncogene 2003; 22:6785-93. [PMID: 14555991 DOI: 10.1038/sj.onc.1206792] [Citation(s) in RCA: 375] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bim is a proapoptotic member of the Bcl-2 family that shares only the BH3 domain with this family. Three Bim proteins Bim-EL, Bim-L and Bim-S are synthesized from the same transcript. We report here that Bim-EL when phosphorylated by Erk1/2 is rapidly degraded via the proteasome pathway. Using different cellular models we evidence that serine 69 is both necessary and sufficient for Erk1/2-mediated phosphorylation and degradation of Bim-EL. In K562 cells, Phorbol 12-myristate 13-acetate activates Erk1/2 and consequently increases Bim-EL phosphorylation and degradation by the proteasome, resulting in cell survival, while the Bcr-Abl inhibitor imatinib abrogates Bim-EL phosphorylation and degradation and induces caspase activation and apoptosis. We also show that Bim-EL(S69G) promotes apoptosis more efficiently than Bim-EL-WT in K562 cells. Altogether, our findings demonstrate that phosphorylation of Bim-EL by Erk1/2 on serine 69 selectively leads to its proteasomal degradation and therefore represents a new and important mechanism of Bim regulation.
Collapse
Affiliation(s)
- Frederic Luciano
- INSERM Unit 526, LNC Label, Faculté de Médecine, 28 Avenue de Valombrose, 06107 Nice Cedex 02, France
| | | | | | | | | | | | | |
Collapse
|
23
|
Mouzakiti A, Packham G. Regulation of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in Burkitt's lymphoma cell lines. Br J Haematol 2003; 122:61-9. [PMID: 12823346 DOI: 10.1046/j.1365-2141.2003.04424.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis in sensitive cells and may be suitable for novel anti-cancer therapies aimed at inducing apoptosis via the activation of TRAIL receptors on malignant cells. Here we have characterized the TRAIL sensitivity of a panel of Burkitt's lymphoma (BL) cell lines. Overall, 5/12 BL cell lines and 1/2 lymphoblastoid cell lines were sensitive to TRAIL-induced apoptosis, although only one BL cell line approached the sensitivity of Jurkat cells, a widely used model for TRAIL-induced apoptosis. Whereas, 4/5 of the Epstein-Barr virus (EBV)-negative cell lines were TRAIL sensitive, only 1/7 EBV-positive BL cell lines were TRAIL sensitive. However, isogenic BL cell lines with different EBV status were not differently sensitive to TRAIL, indicating that EBV is not a major determinant of TRAIL sensitivity. All cell lines expressed the death receptor (DR)5 TRAIL receptor, whereas expression of DR4 was more variable. Differences in the expression of downstream signalling molecules [Fas-associated death domain protein (FADD), caspase 8] and inhibitors [decoy receptor 1 (DcR1), cellular FLICE-like inhibitory protein (c-FLIP)] did not correlate with TRAIL sensitivity. Therefore, a subset of BL cell lines are sensitive to TRAIL-induced apoptosis, however, the molecular mechanism that determines responsiveness remains to be identified.
Collapse
Affiliation(s)
- Amalia Mouzakiti
- Cancer Research UK Oncology Unit, Cancer Sciences Division, School of Medicine, University of Southampton, UK
| | | |
Collapse
|
24
|
Serafeim A, Holder MJ, Grafton G, Chamba A, Drayson MT, Luong QT, Bunce CM, Gregory CD, Barnes NM, Gordon J. Selective serotonin reuptake inhibitors directly signal for apoptosis in biopsy-like Burkitt lymphoma cells. Blood 2003; 101:3212-9. [PMID: 12515726 DOI: 10.1182/blood-2002-07-2044] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the treatment of choice for clinical depression and a range of anxiety-related disorders. They are well tolerated over extended periods with more than 50 million people worldwide benefiting from their use. Here we show that 3 structurally distinct SSRIs--fluoxetine, paroxetine, and citalopram--act directly on Burkitt lymphoma (BL) cells to trigger rapid and extensive programmed cell death. SSRIs unexpectedly stimulated calcium flux, tyrosine phosphorylation, and down-regulation of the c-myc and nm23 genes in Burkitt lymphoma cells remaining faithful to the biopsy phenotype. Resultant SSRI-induced apoptosis was preceded by caspase activation, poly(ADP-ribose) polymerase-1 (PARP-1) cleavage, DNA fragmentation, a loss of mitochondrial membrane potential, and the externalization of phosphatidylserine, and reversed by the overexpression of bcl-2. Normal peripheral blood mononuclear cells and tonsil B cells, whether resting or stimulated into cycle, were largely resistant to SSRI-induced death as were 5 non-BL lymphoid cell lines tested. We discuss these findings within the context of whether the SSRI class of antidepressants could find future application as potential therapeutics for the highly aggressive and-because of its association with AIDS-increasingly more common Burkitt lymphoma.
Collapse
Affiliation(s)
- Adamantios Serafeim
- Medical Research Council Centre for Immune Regulation, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Luciano F, Herrant M, Jacquel A, Ricci JE, Auberger P. The p54 cleaved form of the tyrosine kinase Lyn generated by caspases during BCR-induced cell death in B lymphoma acts as a negative regulator of apoptosis. FASEB J 2003; 17:711-3. [PMID: 12586738 DOI: 10.1096/fj.02-0716fje] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Engagement of the B cell receptor antigen (BCR) triggers apoptosis on immature B cell lines. We report here that BCR triggering leads to caspase activation followed by Lyn cleavage and induction of apoptosis. The cleavage process is mitochondrion-dependent and involves caspases 9 and 7. Stable expression of the cleaved form of Lyn (Lyn-Delta-N) in Ramos B cells impairs BCR-mediated apoptosis as judged by loss of Delta(psi)m, caspase activation and PARP cleavage. Activation of the main survival pathways upon BCR-triggering was unaltered in both cell variants. However, the PI3-K inhibitor Ly294002 resensitizes Lyn-Delta-N cells to apoptosis. Selected cDNA expression arrays revealed that anti-IgM modulates the expression of approximately 20 genes in both cell variants. Among them, only c-Myc was found to be differentially regulated, which suggests a role for c-Myc in the B cell apoptotic response. Interestingly, c-Myc expression decreased more rapidly in Lyn-Delta-N compared with Lyn-WT cells during the first hours of anti-IgM stimulation. Nevertheless, rapid down-regulation of c-Myc following BCR engagement seems to correlate with the resistance of B cells to apoptosis. Thus, the soluble form of Lyn generated by caspases following BCR triggering acts as an inhibitor of B lymphocyte death likely through the modulation of c-Myc expression.
Collapse
Affiliation(s)
- Frederic Luciano
- INSERM U526 Activation des Cellules Hematopoietiques, Physiopathologie de la Survie et de la Mort Cellulaires et Infections Virales, Equipe Labellisée Ligue Nationale contre le Cancer, IFR50, Faculté de Médecine, 06107 Nice-Cédex 2, France
| | | | | | | | | |
Collapse
|
26
|
Abstract
AIM: To observe the localization of TRAIL/TRAIR (DR4, DR5, DcR1, DcR2) in the fetal pancreas.
METHODS: Fetal pancreas of 32 wk of pregnancy were obtained from induced abortions, embedded in paraffin, and 4-μm sections were prepared. The localization of TRAIL/TRAILR in fetal pancreas was investigated by fluorescence immunohistochemical method combined with laser scanning confocal microscopy.
RESULTS: TRAIL immunoreactive cells were mainly located on the periphery of the pancreas islets. There were a few DcR1 and DcR2 positive cells whereas there were no immunoreactive cells of DR4 and DR5 in the pancreas islets. In the acini and the ducts of the exocrine pancreas there were no TRAIL/TRAILR immunoreactive cells.
CONCLUSION: This study not only describes the distribution of TRAIL/TRAILR in the fetal pancreas, but also provides a morphological basis for deducing the function of TRAIL/TRAILR in pancreas, suggesting that in normal pancreatic islets, the pancreatic cells are resistant towards apoptosis too.
Collapse
Affiliation(s)
- Li-Hua Chen
- Department of Immunology, the Fourth Military Medical University, 17 West Changle Road, Xi'an 710032, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|