1
|
Demir I, Pehlivan S, Okan V, Sahin HH, Durusoy SS, Serin I, Oyaci Y, Pehlivan M. Effect of the uncoupling protein-2 (UCP-2) and nuclear receptor subfamily 3 group C member 1 (NR3C1) genes on treatment efficacy and survival in patients with multiple myeloma: a single-center study. BMC Res Notes 2021; 14:346. [PMID: 34481515 PMCID: PMC8418283 DOI: 10.1186/s13104-021-05758-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/23/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Studies on the genetic background of patients with multiple myeloma (MM) have been increasing; two important factors considered in such works are uncoupling protein-2 (UCP-2) and nuclear receptor subfamily 3 group C member 1 (NR3C1). We aim to reveal the association of MM with NR3C1 and UCP-2 gene polymorphisms. In this prospective study, 200 patients diagnosed between January 2009 and 2018 and 200 healthy individuals were included. For patients who had undergone autologous stem cell transplantation and control subjects, we statistically compared the CC, GC, and GG genotypes and the C and G alleles of the NR3C1 gene, as well as the AA, AG, and GG genotypes and the A and G alleles of the UCP-2 gene. RESULTS While the AA genotype was significantly more common in the MM group (p = 0.001), the GG genotype was significantly more common in the control group (p = 0.016). Overall survival was found to be significantly shorter in patients with the UCP-2 GG genotype (p = 0.034). It was also found that having the GG genotype of the UCP-2 gene was a 2.48-fold risk factor for mortality. The fact that overall survival is significantly shorter in MM patients with the UCP-2 GG genotype and its definition as a risk factor for mortality have been put forward for the first time in the literature.
Collapse
Affiliation(s)
- Ilknur Demir
- Department of Internal Medicine, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Sacide Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Vahap Okan
- Department of Hematology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | | | - Salih Sertaç Durusoy
- Department of Hematology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Istemi Serin
- Department of Hematology, University of Health Sciences, Istanbul Training and Research Hospital, Org. Nafiz Gurman Cad., Fatih, 34098, Istanbul, Turkey.
| | - Yasemin Oyaci
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mustafa Pehlivan
- Department of Hematology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey.,Department of Hematology, University of Health Sciences, Istanbul Training and Research Hospital, Org. Nafiz Gurman Cad., Fatih, 34098, Istanbul, Turkey
| |
Collapse
|
2
|
Glucocorticoid-Induced Leucine Zipper (GILZ) in Cardiovascular Health and Disease. Cells 2021; 10:cells10082155. [PMID: 34440924 PMCID: PMC8394287 DOI: 10.3390/cells10082155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 01/13/2023] Open
Abstract
Glucocorticoids (GCs) are essential in regulating functions and homeostasis in many biological systems and are extensively used to treat a variety of conditions associated with immune/inflammatory processes. GCs are among the most powerful drugs for the treatment of autoimmune and inflammatory diseases, but their long-term usage is limited by severe adverse effects. For this reason, to envision new therapies devoid of typical GC side effects, research has focused on expanding the knowledge of cellular and molecular effects of GCs. GC-induced leucine zipper (GILZ) is a GC-target protein shown to mediate several actions of GCs, including inhibition of the NF-κB and MAPK pathways. GILZ expression is not restricted to immune cells, and it has been shown to play a regulatory role in many organs and tissues, including the cardiovascular system. Research on the role of GILZ on endothelial cells has demonstrated its ability to modulate the inflammatory cascade, resulting in a downregulation of cytokines, chemokines, and cellular adhesion molecules. GILZ also has the capacity to protect myocardial cells, as its deletion makes the heart, after a deleterious stimulus, more susceptible to apoptosis, immune cell infiltration, hypertrophy, and impaired function. Despite these advances, we have only just begun to appreciate the relevance of GILZ in cardiovascular homeostasis and dysfunction. This review summarizes the current understanding of the role of GILZ in modulating biological processes relevant to cardiovascular biology.
Collapse
|
3
|
Spaan I, Timmerman LM, Kimman T, Slomp A, Cuenca M, van Nieuwenhuijzen N, Moesbergen LM, Minnema MC, Raymakers RA, Peperzak V. Direct P70S6K1 inhibition to replace dexamethasone in synergistic combination with MCL-1 inhibition in multiple myeloma. Blood Adv 2021; 5:2593-2607. [PMID: 34152396 PMCID: PMC8270664 DOI: 10.1182/bloodadvances.2020003624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Novel combination therapies have markedly improved the lifespan of patients with multiple myeloma (MM), but drug resistance and disease relapse remain major clinical problems. Dexamethasone and other glucocorticoids are a cornerstone of conventional and new combination therapies for MM, although their use is accompanied by serious side effects. We aimed to uncover drug combinations that act in synergy and, as such, allow reduced dosing while remaining effective. Dexamethasone and the myeloid cell leukemia 1 (MCL-1) inhibitor S63845 (MCL-1i) proved the most potent combination in our lethality screen and induced apoptosis of human myeloma cell lines (HMCLs) that was 50% higher compared with an additive drug effect. Kinome analysis of dexamethasone-treated HMCLs revealed a reduction in serine/threonine peptide phosphorylation, which was predicted to result from reduced Akt activity. Biochemical techniques showed no dexamethasone-induced effects on FOXO protein or GSK3 but did show a 50% reduction in P70S6K phosphorylation, downstream of the Akt-mTORC1 axis. Replacing dexamethasone by the P70S6K1 isoform-specific inhibitor PF-4708671 (S6K1i) revealed similar and statistically significant synergistic apoptosis of HMCLs in combination with MCL-1i. Interestingly, apoptosis induced by the P70S6K1i and MCL-1i combination was more-than-additive in all 9 primary MM samples tested; this effect was observed for 6 of 9 samples with the dexamethasone and MCL-1i combination. Toxicity on stem and progenitor cell subsets remained minimal. Combined, our results show a strong rationale for combination treatments using the P70S6K inhibitor in MM. Direct and specific inhibition of P70S6K may also provide a solution for patients ineligible or insensitive to dexamethasone or other glucocorticoids.
Collapse
Affiliation(s)
| | | | | | | | | | - Niels van Nieuwenhuijzen
- Center for Translational Immunology and
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Reinier A Raymakers
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
4
|
Bӧhm JW, Sia KCS, Jones C, Evans K, Mariana A, Pang I, Failes T, Zhong L, Mayoh C, Landman R, Collins R, Erickson SW, Arndt G, Raftery MJ, Wilkins MR, Norris MD, Haber M, Marshall GM, Lock RB. Combination efficacy of ruxolitinib with standard-of-care drugs in CRLF2-rearranged Ph-like acute lymphoblastic leukemia. Leukemia 2021; 35:3101-3112. [PMID: 33895784 DOI: 10.1038/s41375-021-01248-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 11/09/2022]
Abstract
Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk ALL subtype with high rates of relapse and poor patient outcome. Activating mutations affecting components of the JAK-STAT signaling pathway occur in the majority of Ph-like ALL cases. The use of JAK inhibitors represents a potential treatment option for Ph-like ALL, although we and others have shown that CRLF2-rearranged Ph-like ALL responds poorly to single-agent JAK inhibitors in the preclinical setting. Therefore, the aim of this study was to identify effective combination treatments against CRLF2-rearranged Ph-like ALL, and to elucidate the underlying mechanisms of synergy. We carried out a series of high-throughput combination drug screenings and found that ruxolitinib exerted synergy with standard-of-care drugs used in the treatment of ALL. In addition, we investigated the molecular effects of ruxolitinib on Ph-like ALL by combining mass spectrometry phosphoproteomics with gene expression analysis. Based on these findings, we conducted preclinical in vivo drug testing and demonstrated that ruxolitinib enhanced the in vivo efficacy of an induction-type regimen consisting of vincristine, dexamethasone, and L-asparaginase in 2/3 CRLF2-rearranged Ph-like ALL xenografts. Overall, our findings support evaluating the addition of ruxolitinib to conventional induction regimens for the treatment of CRLF2-rearranged Ph-like ALL.
Collapse
Affiliation(s)
- Julia W Bӧhm
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Keith C S Sia
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Connor Jones
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Kathryn Evans
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Anna Mariana
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Ignatius Pang
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Tim Failes
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, UNSW Sydney, Sydney, NSW, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | | | | | | | - Greg Arndt
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Mark J Raftery
- Bioanalytical Mass Spectrometry Facility, UNSW Sydney, Sydney, NSW, Australia
| | - Marc R Wilkins
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Glenn M Marshall
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, School of Women's and Children's Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Chen S, Du Y, Xu B, Li Q, Yang L, Jiang Z, Zeng Z, Chen L. Vaccinia-related kinase 2 blunts sorafenib's efficacy against hepatocellular carcinoma by disturbing the apoptosis-autophagy balance. Oncogene 2021; 40:3378-3393. [PMID: 33875785 DOI: 10.1038/s41388-021-01780-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy with limited treatment options. Sorafenib is the only Food and Drug Administration (FDA)-approved first-line targeted drug for the treatment of advanced HCC. However, its effect on patient survival is limited. Recently, studies have demonstrated that the imbalance between apoptosis and autophagy plays a critical role in chemoresistance, and it is hypothesised that restoring the balance between these processes is a potential treatment strategy for improving chemoresistance in cancer. However, there is currently no evidence supporting this hypothesis. We aimed to investigate if vaccinia-related kinase 2 (VRK2), a serine/threonine protein kinase, confers sorafenib resistance in HCC cells. Here, we found that VRK2 was enriched in sorafenib-resistant HCC cells and patient-derived xenografts. Both in vivo and in vitro evidences showed that VRK2 blunts the efficacy of sorafenib against hepatocellular carcinoma by disturbing the balance between apoptosis and autophagy. Mechanistically, VRK2 promotes the phosphorylation of Bcl-2 by activating JNK1/MAPK8, thereby enhancing the dissociation of Bcl-2 from Beclin-1 and promoting the formation of the Beclin-1-Atg14-Vps34 complex, which facilitates autophagy. Furthermore, VRK2-induced phosphorylation of Bcl-2 promotes the interaction of Bcl-2 with BAX, thereby inhibiting apoptosis. In conclusion, targeting VRK2 for modulation of the balance between autophagy and apoptosis may be a novel strategy for overcoming sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Sisi Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunyan Du
- Department of Medical, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Bin Xu
- Department of Burns, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qing Li
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Le Yang
- Department of Pharmacy, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zi Jiang
- Department of Pharmacy, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhaoxia Zeng
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
6
|
Sciarrillo R, Wojtuszkiewicz A, Assaraf YG, Jansen G, Kaspers GJL, Giovannetti E, Cloos J. The role of alternative splicing in cancer: From oncogenesis to drug resistance. Drug Resist Updat 2020; 53:100728. [PMID: 33070093 DOI: 10.1016/j.drup.2020.100728] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Alternative splicing is a tightly regulated process whereby non-coding sequences of pre-mRNA are removed and protein-coding segments are assembled in diverse combinations, ultimately giving rise to proteins with distinct or even opposing functions. In the past decade, whole genome/transcriptome sequencing studies revealed the high complexity of splicing regulation, which occurs co-transcriptionally and is influenced by chromatin status and mRNA modifications. Consequently, splicing profiles of both healthy and malignant cells display high diversity and alternative splicing was shown to be widely deregulated in multiple cancer types. In particular, mutations in pre-mRNA regulatory sequences, splicing regulators and chromatin modifiers, as well as differential expression of splicing factors are important contributors to cancer pathogenesis. It has become clear that these aberrations contribute to many facets of cancer, including oncogenic transformation, cancer progression, response to anticancer drug treatment as well as resistance to therapy. In this respect, alternative splicing was shown to perturb the expression a broad spectrum of relevant genes involved in drug uptake/metabolism (i.e. SLC29A1, dCK, FPGS, and TP), activation of nuclear receptor pathways (i.e. GR, AR), regulation of apoptosis (i.e. MCL1, BCL-X, and FAS) and modulation of response to immunotherapy (CD19). Furthermore, aberrant splicing constitutes an important source of novel cancer biomarkers and the spliceosome machinery represents an attractive target for a novel and rapidly expanding class of therapeutic agents. Small molecule inhibitors targeting SF3B1 or splice factor kinases were highly cytotoxic against a wide range of cancer models, including drug-resistant cells. Importantly, these effects are enhanced in specific cancer subsets, such as splicing factor-mutated and c-MYC-driven tumors. Furthermore, pre-clinical studies report synergistic effects of spliceosome modulators in combination with conventional antitumor agents. These strategies based on the use of low dose splicing modulators could shift the therapeutic window towards decreased toxicity in healthy tissues. Here we provide an extensive overview of the latest findings in the field of regulation of splicing in cancer, including molecular mechanisms by which cancer cells harness alternative splicing to drive oncogenesis and evade anticancer drug treatment as well as splicing-based vulnerabilities that can provide novel treatment opportunities. Furthermore, we discuss current challenges arising from genome-wide detection and prediction methods of aberrant splicing, as well as unravelling functional relevance of the plethora of cancer-related splicing alterations.
Collapse
Affiliation(s)
- Rocco Sciarrillo
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Department of Pediatric Oncology, Emma's Children's Hospital, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Anna Wojtuszkiewicz
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Gerrit Jansen
- Amsterdam Immunology and Rheumatology Center, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology, Emma's Children's Hospital, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands; Fondazione Pisana per la Scienza, Pisa, Italy
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
7
|
Clarisse D, Offner F, De Bosscher K. Latest perspectives on glucocorticoid-induced apoptosis and resistance in lymphoid malignancies. Biochim Biophys Acta Rev Cancer 2020; 1874:188430. [PMID: 32950642 DOI: 10.1016/j.bbcan.2020.188430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/13/2020] [Accepted: 09/14/2020] [Indexed: 02/09/2023]
Abstract
Glucocorticoids are essential drugs in the treatment protocols of lymphoid malignancies. These steroidal hormones trigger apoptosis of the malignant cells by binding to the glucocorticoid receptor (GR), which is a member of the nuclear receptor superfamily. Long term glucocorticoid treatment is limited by two major problems: the development of glucocorticoid-related side effects, which hampers patient quality of life, and the emergence of glucocorticoid resistance, which is a gradual process that is inevitable in many patients. This emphasizes the need to reevaluate and optimize the widespread use of glucocorticoids in lymphoid malignancies. To achieve this goal, a deep understanding of the mechanisms governing glucocorticoid responsiveness is required, yet, a recent comprehensive overview is currently lacking. In this review, we examine how glucocorticoids mediate apoptosis by detailing GR's genomic and non-genomic action mechanisms in lymphoid malignancies. We continue with a discussion of the glucocorticoid-related problems and how these are intertwined with one another. We further zoom in on glucocorticoid resistance by critically analyzing the plethora of proposed mechanisms and highlighting therapeutic opportunities that emerge from these studies. In conclusion, early detection of glucocorticoid resistance in patients remains an important challenge as this would result in a timelier treatment reorientation and reduced glucocorticoid-instigated side effects.
Collapse
Affiliation(s)
- Dorien Clarisse
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Fritz Offner
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Karolien De Bosscher
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
8
|
Zhu X, Wu S, Zeng W, Chen X, Zheng T, Ren J, Ke M. Protective Effects of Rapamycin on Trabecular Meshwork Cells in Glucocorticoid-Induced Glaucoma Mice. Front Pharmacol 2020; 11:1006. [PMID: 32714192 PMCID: PMC7344368 DOI: 10.3389/fphar.2020.01006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid-induced glaucoma (GIG) is a chronic optic neuropathy caused by systemic or topical glucocorticoid (GC) treatment, which could eventually lead to permanent vision loss. To investigate the protective effects of rapamycin (RAP) on the trabecular cells during the development of GIG in mice, the effects of RAP on intraocular pressure (IOP), trabecular ultrastructure, and retinal ganglion cells (RGCs) were examined in C57BL/6J female mice treated with dexamethasone acetate (Dex-Ace). The expression of α-actin in trabecular tissue was detected by immunofluorescence, and the autophagic activity of trabecular cells and the expression of GIG-related myocilin and α-actin were detected by immunoblotting. Our results indicated that Dex-Ace significantly increased IOP at the end of the third week (p < 0.05), while RAP treatment neutralized this elevation of IOP by Dex-Ace. Dex-Ace treatment significantly decreased the RGC numbers (p < 0.05), while synchronous RAP treatment kept the number comparable to control. The outer sheath of elastic fibers became thicker and denser, and the mitochondria of lesions increased in Dex-Ace-treated groups at 4 weeks, while no significant change was observed in the RAP-treated trabecular tissues. Dex-Ace induced myocilin, α-actin, Beclin-1, and LC3-II/LC-I ratio, and lowered p62, while synchronous RAP treatment further activated autophagy and neutralized the induction of myocilin and α-actin. Our studies suggested that RAP protected trabecular meshwork cells by further inducing autophagy way from damages of GC treatment.
Collapse
Affiliation(s)
- Xiaolu Zhu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shengyu Wu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Zeng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Chen
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Zheng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiangbo Ren
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Sciarrillo R, Wojtuszkiewicz A, Kooi IE, Leon LG, Sonneveld E, Kuiper RP, Jansen G, Giovannetti E, Kaspers GJ, Cloos J. Glucocorticoid Resistant Pediatric Acute Lymphoblastic Leukemia Samples Display Altered Splicing Profile and Vulnerability to Spliceosome Modulation. Cancers (Basel) 2020; 12:cancers12030723. [PMID: 32204435 PMCID: PMC7140081 DOI: 10.3390/cancers12030723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 12/01/2022] Open
Abstract
Glucocorticoid (GC) resistance is a crucial determinant of inferior response to chemotherapy in pediatric acute lymphoblastic leukemia (ALL); however, molecular mechanisms underlying this phenomenon are poorly understood. Deregulated splicing is a common feature of many cancers, which impacts drug response and constitutes an attractive therapeutic target. Therefore, the aim of the current study was to characterize global splicing profiles associated with GC resistance and determine whether splicing modulation could serve as a novel therapeutic option for GC-resistant patients. To this end, 38 primary ALL samples were profiled using RNA-seq-based differential splicing analysis. The impact of splicing modulators was investigated in GC-resistant leukemia cell lines and primary leukemic specimens. Our findings revealed, for the first time, markedly distinct splicing landscapes in ALL samples of B-cell precursor (BCP)-ALL and T-ALL lineages. Differential splicing events associated with GC resistance were involved in RNA processing, a direct response to GCs, survival signaling, apoptosis, cell cycle regulation and energy metabolism. Furthermore, our analyses showed that GC-resistant ALL cell lines and primary samples are sensitive to splicing modulation, alone and in combination with GC. Together, these findings suggest that aberrant splicing is associated with GC resistance and splicing modulators deserve further interest as a novel treatment option for GC-resistant patients.
Collapse
Affiliation(s)
- Rocco Sciarrillo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Departments of Pediatric Oncology, Hematology and Medical Oncology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Anna Wojtuszkiewicz
- Amsterdam UMC, Vrije Universiteit Amsterdam, Departments of Pediatric Oncology and Hematology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| | - Irsan E. Kooi
- Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Department of Clinical Genetics, 1081 HV Amsterdam, The Netherlands
| | - Leticia G. Leon
- Erasmus MC, University Medical Center Rotterdam, Department of Immunology, 3000 CA Rotterdam, The Netherlands
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, 3584 CX Utrecht, The Netherlands
| | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, 3584 CX Utrecht, The Netherlands
| | - Gerrit Jansen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Immunology and Rheumatology Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, 56017 San Giuliano Terme (Pisa), Italy
| | - Gertjan J.L. Kaspers
- Princess Máxima Center for Pediatric Oncology, 3584 CX Utrecht, The Netherlands
- Emma’s Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1081 HV Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
10
|
Tamayo-Orbegozo E, Amo L, Díez-García J, Amutio E, Riñón M, Alonso M, Arana P, Maruri N, Larrucea S. Emerging Role of Podocalyxin in the Progression of Mature B-Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2020; 12:cancers12020396. [PMID: 32046309 PMCID: PMC7072361 DOI: 10.3390/cancers12020396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Mature B-cell non-Hodgkin lymphoma (B-NHL) constitutes a group of heterogeneous malignant lymphoproliferative diseases ranging from indolent to highly aggressive forms. Although the survival after chemo-immunotherapy treatment of mature B-NHL has increased over the last years, many patients relapse or remain refractory due to drug resistance, presenting an unfavorable prognosis. Hence, there is an urgent need to identify new prognostic markers and therapeutic targets. Podocalyxin (PODXL), a sialomucin overexpressed in a variety of tumor cell types and associated with their aggressiveness, has been implicated in multiple aspects of cancer progression, although its participation in hematological malignancies remains unexplored. New evidence points to a role for PODXL in mature B-NHL cell proliferation, survival, migration, drug resistance, and metabolic reprogramming, as well as enhanced levels of PODXL in mature B-NHL. Here, we review the current knowledge on the contribution of PODXL to tumorigenesis, highlighting and discussing its role in mature B-NHL progression.
Collapse
Affiliation(s)
- Estíbaliz Tamayo-Orbegozo
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (E.T.-O.); (L.A.)
| | - Laura Amo
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (E.T.-O.); (L.A.)
| | - Javier Díez-García
- Microscopy Facility, Biocruces Bizkaia Health Research Institute, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain;
| | - Elena Amutio
- Blood Cancer Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain;
| | - Marta Riñón
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (M.R.); (M.A.); (P.A.); (N.M.)
| | - Marta Alonso
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (M.R.); (M.A.); (P.A.); (N.M.)
| | - Paula Arana
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (M.R.); (M.A.); (P.A.); (N.M.)
| | - Natalia Maruri
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (M.R.); (M.A.); (P.A.); (N.M.)
| | - Susana Larrucea
- Regulation of the Immune System Group, Biocruces Bizkaia Health Research Institute, Hospital Universitario Cruces, Plaza de Cruces 12, 48903 Barakaldo, Bizkaia, Spain; (M.R.); (M.A.); (P.A.); (N.M.)
- Correspondence:
| |
Collapse
|
11
|
Petty AJ, Yang Y. Tumor-Associated Macrophages in Hematologic Malignancies: New Insights and Targeted Therapies. Cells 2019; 8:cells8121526. [PMID: 31783588 PMCID: PMC6952752 DOI: 10.3390/cells8121526] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
The growth of hematologic malignant cells can be facilitated by other non-tumor cells within the same microenvironment, including stromal, vascular, immune and mesenchymal stem cells. Macrophages are an integral part of the human innate immune system and the tumor microenvironment. Complex interplays between the malignant hematologic cells and the infiltrating macrophages promote the formation of leukemia, lymphoma or myeloma-associated macrophages. These pro-tumorigenic macrophages in turn play an important part in facilitating tumor growth, metastasis and chemotherapeutic resistance. Previous reports have highlighted the association between tumor-associated macrophages (TAMs) and disease progression in hematologic malignancies. This review summarizes the role of TAMs in different subtypes of leukemia, lymphoma and myeloma, focusing on new insights and targeted therapies.
Collapse
Affiliation(s)
- Amy J. Petty
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA;
- Division of Hematology, The Ohio State University Wexner Medical Center, 508 BRT, 460 W 12th Avenue, Columbus, OH 43210, OH, USA
| | - Yiping Yang
- Division of Hematology, The Ohio State University Wexner Medical Center, 508 BRT, 460 W 12th Avenue, Columbus, OH 43210, OH, USA
- Correspondence: ; Tel.: +1-(614)-685-0643; Fax: +1-(614)-293-7526
| |
Collapse
|
12
|
Liu B, Zhang TN, Knight JK, Goodwin JE. The Glucocorticoid Receptor in Cardiovascular Health and Disease. Cells 2019; 8:cells8101227. [PMID: 31601045 PMCID: PMC6829609 DOI: 10.3390/cells8101227] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
The glucocorticoid receptor is a member of the nuclear receptor family that controls many distinct gene networks, governing various aspects of development, metabolism, inflammation, and the stress response, as well as other key biological processes in the cardiovascular system. Recently, research in both animal models and humans has begun to unravel the profound complexity of glucocorticoid signaling and convincingly demonstrates that the glucocorticoid receptor has direct effects on the heart and vessels in vivo and in vitro. This research has contributed directly to improving therapeutic strategies in human disease. The glucocorticoid receptor is activated either by the endogenous steroid hormone cortisol or by exogenous glucocorticoids and acts within the cardiovascular system via both genomic and non-genomic pathways. Polymorphisms of the glucocorticoid receptor are also reported to influence the progress and prognosis of cardiovascular disease. In this review, we provide an update on glucocorticoid signaling and highlight the critical role of this signaling in both physiological and pathological conditions of the cardiovascular system. With increasing in-depth understanding of glucocorticoid signaling, the future is promising for the development of targeted glucocorticoid treatments and improved clinical outcomes.
Collapse
Affiliation(s)
- Bing Liu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Tie-Ning Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jessica K Knight
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
13
|
Zeng W, Wang W, Wu S, Zhu X, Zheng T, Chen X, Ren J, Gong Y, Ke M. Mitochondria and Autophagy Dysfunction in Glucocorticoid-Induced Ocular Hypertension/Glaucoma Mice Model. Curr Eye Res 2019; 45:190-198. [PMID: 31425668 DOI: 10.1080/02713683.2019.1657462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose: This study is aimed to investigate the effects of periocular steroids induction on intraocular pressure (IOP), retinal ganglion cells (RGCs) and trabecular meshwork (TM) ultrastructure in glucocorticoid-induced ocular hypertension mice model.Materials and Methods: Dexamethasone-21-acetate (Dex-Ace) was administered through periocular conjunctival fornix injection every 3 days in C57BL/6J mice. Intraocular pressure was measured weekly by rebound tonometry. RGCs were examined with immunofluorescent staining of BRN3a at week 1, 4, and 8. TM morphology was visualized with electron microscopy. Autophagy was evaluated with immunoblotting in TM tissues.Results: Dex-Ace rapidly and significantly induced IOP, which peaked at week 4. The absolute increase in IOP in the Dex-Ace-treated mice was 8.1 ± 1.4 mmHg, a 60% induction (p < .0001) compared with that in the vehicle-treated mice. The IOP sustained a higher level in the Dex-Ace group from week 4 to week 8. Dex-Ace treatment decreased the number of RGCs in a time-dependent manner, suggesting that high IOP resulted in optic neuropathy. In addition, Dex-Ace thickened trabecular beams and decreased intertrabecular spaces, with marked accumulation of fibrillar and amorphous granular extracellular material. Moreover, Dex-Ace induced swollen and elongated mitochondria in TM cells. The average mitochondria area was 0.090 ± 0.044 µm2 in the vehicle-treated mice, and increased to 0.161 ± 0.094 µm2 (p < .0001), 0.121 ± 0.029 µm2 (p = .0223) and 0.171 ± 0.076 µm2 (p < .0001) in the Dex-Ace-treated mice at weeks 1, 4 and 8, respectively. Autophagy was also increased by Dex-Ace treatment, indicating by the upregulation of LC3-I, LC3-II and beclin-1, and downregulation of p62.Conclusion: Dex-Ace administration decreased RGCs and changed TM ultrastructure, mimicking hallmarks of human glucocorticoid-induced glaucoma (GIG). In addition, mitochondria and autophagy dysfunction suggested abnormal energy metabolism in TM cells, which warranted further study to fully elucidate the pathogenesis of GIG.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenhuan Wang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shengyu Wu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolu Zhu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tian Zheng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Chen
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiangbo Ren
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Ibrutinib reprograms the glucocorticoid receptor in chronic lymphocytic leukemia cells. Leukemia 2019; 33:1650-1662. [PMID: 30696950 DOI: 10.1038/s41375-019-0381-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/12/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
Glucocorticoid (GC) receptor (GR) phosphorylation and signature genes were studied in chronic lymphocytic leukemia (CLL) cells to help place GCs within modern treatment algorithms. In contrast to normal B and T cells, transcription of GC-regulated genes was not rhythmic and the synthetic GC dexamethasone (DEX) could not inhibit toll-like receptor (TLR)-responses in CLL cells. This intrinsic GC-resistance was associated with aberrant GR-phosphorylation on activating Ser211 and inhibitory Ser226 sites. Ibrutinib increased transcription of the GR-signature gene GILZ in circulating CLL cells along with GR(pS211)/GR(pS226) ratios and lytic sensitivity to DEX that were not reversed by the competitive antagonist mifepristone in vitro. However, ibrutinib could not improve GR-responses in circulating CLL cells activated with IL2 and TLR7/8 agonists to mimic conditions in pseudofollicle microenvironments. Addition of the janus kinase inhibitor ruxolitinib to block ibrutinib-insensitive signals increased GILZ transcription in pseudofollicle conditions in vitro and in a clinical trial (NCT02912754), and also increased GR(S211)/GR(S226) ratios and DEX-mediated killing in patient samples in vitro. These observations suggest that intrinsic resistance to endogenous GCs is characteristic of CLL cells and ibrutinib may help increase the therapeutic activity of GCs by non-canonical activation of GR.
Collapse
|
15
|
Webb MS, Miller AL, Howard TL, Johnson BH, Chumakov S, Fofanov Y, Nguyen-Vu T, Lin CY, Thompson EB. Sequential gene regulatory events leading to glucocorticoid-evoked apoptosis of CEM human leukemic cells:interactions of MAPK, MYC and glucocorticoid pathways. Mol Cell Endocrinol 2018; 471:118-130. [PMID: 29596968 PMCID: PMC6075652 DOI: 10.1016/j.mce.2018.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 02/13/2018] [Accepted: 03/07/2018] [Indexed: 12/22/2022]
Abstract
Gene expression responses to glucocorticoid (GC) in the hours preceding onset of apoptosis were compared in three clones of human acute lymphoblastic leukemia CEM cells. Between 2 and 20h, all three clones showed increasing numbers of responding genes. Each clone had many unique responses, but the two responsive clones showed a group of responding genes in common, different from the resistant clone. MYC levels and the balance of activities between the three major groups of MAPKs are known important regulators of glucocorticoid-driven apoptosis in several lymphoid cell systems. Common to the two sensitive clones were changed transcript levels from genes that decrease amounts or activity of anti-apoptotic ERK/MAPK1 and JNK2/MAPK9, or of genes that increase activity of pro-apoptotic p38/MAPK14. Down-regulation of MYC and several MYC-regulated genes relevant to MAPKs also occurred in both sensitive clones. Transcriptomine comparisons revealed probable NOTCH-GC crosstalk in these cells.
Collapse
Affiliation(s)
- M S Webb
- Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555, USA
| | - A L Miller
- Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555, USA
| | - T L Howard
- Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555, USA
| | - B H Johnson
- Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555, USA
| | - S Chumakov
- Dept. of Computer Science, Dept. of Physics, University of Guadalahara, Gaudalahara, Jalisco, Mexico
| | - Y Fofanov
- Dept. of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston TX 77555, USA
| | - T Nguyen-Vu
- Center for Nuclear Receptors & Cell Signaling, Dept. of Biology & Biochemistry, University of Houston, Houston TX 77204, USA
| | - C Y Lin
- Center for Nuclear Receptors & Cell Signaling, Dept. of Biology & Biochemistry, University of Houston, Houston TX 77204, USA
| | - E B Thompson
- Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston TX 77555, USA; Center for Nuclear Receptors & Cell Signaling, Dept. of Biology & Biochemistry, University of Houston, Houston TX 77204, USA.
| |
Collapse
|
16
|
Cabezas M, Camós M, Rives S, Garcia-Orad A, Lopez-Lopez E, Dapena JL, Caballín MR, Armengol G. Impact of polymorphisms in apoptosis-related genes on the outcome of childhood acute lymphoblastic leukaemia. Br J Haematol 2018; 185:159-162. [PMID: 29808934 DOI: 10.1111/bjh.15415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Maria Cabezas
- Unit of Biological Anthropology, Department of Animal Biology, Plant Biology and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Mireia Camós
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Catalonia, Spain.,Haematology Laboratory, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Catalonia, Spain.,National Biomedical Research Institute on Rare Diseases (CIBER ER), Instituto de Salud Carlos III, Madrid, Spain
| | - Susana Rives
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Catalonia, Spain.,National Biomedical Research Institute on Rare Diseases (CIBER ER), Instituto de Salud Carlos III, Madrid, Spain.,Paediatric Haematology and Oncology Department, Hospital Sant Joan de Déu Barcelona, Esplugues de Llobregat, Catalonia, Spain
| | - Africa Garcia-Orad
- Department of Genetics, Physic Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Elixabet Lopez-Lopez
- Department of Genetics, Physic Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, Bizkaia, Spain
| | - Josep L Dapena
- Service of Paediatric Haematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain
| | - María R Caballín
- Unit of Biological Anthropology, Department of Animal Biology, Plant Biology and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Gemma Armengol
- Unit of Biological Anthropology, Department of Animal Biology, Plant Biology and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| |
Collapse
|
17
|
Ouimet M, Drouin S, Lajoie M, Caron M, St-Onge P, Gioia R, Richer C, Sinnett D. A childhood acute lymphoblastic leukemia-specific lncRNA implicated in prednisolone resistance, cell proliferation, and migration. Oncotarget 2018; 8:7477-7488. [PMID: 27980230 PMCID: PMC5352336 DOI: 10.18632/oncotarget.13936] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 11/25/2022] Open
Abstract
Childhood acute lymphoblastic leukemia (cALL) is the most common pediatric cancer and, despite an 85% cure rate, still represents a major cause of disease-related death in children. Recent studies have implicated long non-coding RNAs (lncRNAs) in cALL etiology, progression, and treatment response. However, barring some exceptions little is known about the functional impact of lncRNAs on cancer biology, which limits their potential as potential therapeutic targets. We wanted to investigate the functional role of lncRNAs identified as specifically overexpressed in pre-B cALL by whole-transcriptome sequencing. Here we report five lncRNAs specifically upregulated in pre-B cALL that had significant impacts on cancer hallmark traits such as cell proliferation, migration, apoptosis, and treatment response. In particular, silencing of the RP11-137H2.4 lncRNA effectively restored normal glucocorticoid (GC) response in a GC-resistant pre-B cALL cell line and specifically modulated expression of members of both the NRAS/BRAF/NF-?B MAPK cascade and cell cycle pathways. Since GC form the cornerstone of cALL chemotherapy and resistance in cALL confers a dismal prognosis, characterizing RP11-137H2.4sexact role and function in this process will be critical to the development of new therapeutic approaches to overcome GC resistance in children treated for cALL.
Collapse
Affiliation(s)
- Manon Ouimet
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Simon Drouin
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Mathieu Lajoie
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Maxime Caron
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Pascal St-Onge
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Romain Gioia
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Chantal Richer
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
18
|
Tsubaki M, Takeda T, Tomonari Y, Mashimo K, Koumoto YI, Hoshida S, Itoh T, Imano M, Satou T, Sakaguchi K, Nishida S. The MIP-1α autocrine loop contributes to decreased sensitivity to anticancer drugs. J Cell Physiol 2017; 233:4258-4271. [PMID: 29057477 DOI: 10.1002/jcp.26245] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 10/18/2017] [Indexed: 01/04/2023]
Abstract
Several autocrine soluble factors, including macrophage inflammatory protein-1α (MIP-1α), tumor necrosis factor-α, and hepatocyte growth factor, promote cell survival and growth in multiple myeloma (MM) cells. We hypothesized that inhibition of the MIP-1α autocrine loop may enhance the cytotoxic effect of anticancer drugs in MM cell lines. In the present study, an MIP-1α neutralizing antibody suppressed cell proliferation and enhanced the cytotoxic effect of melphalan or bortezomib on MM cells. In addition, melphalan resistance cells (RPMI8226/L-PAM and HS-sultan/L-PAM cells) secreted MIP-1α and neutralizing antibody of MIP-1α partially overcame melphalan resistance. Moreover, combination treatment with MIP-1α neutralizing antibody and melphalan or bortezomib inhibited extracellular signal regulated kinase 1/2 (ERK1/2), Akt, and mammalian target of rapamycin (mTOR) activation, Bcl-2, Bcl-xL, and Survivin expression, and upregulated the expression of Bim and cleaved Poly (ADP-ribose) polymerase (PARP). Treatment of IM9 cells with MIP-1α siRNA suppressed the activation of ERK1/2, Akt, and mTOR, and enhanced the cytotoxic effect of melphalan and bortezomib. These results indicate that MIP-1α neutralizing antibodies or MIP-1α siRNA enhance the cytotoxic effect of melphalan and bortezomib by suppressing the chemokine receptor/ERK and chemokine receptor/Akt/mTOR pathways. The inhibition of MIP-1α may thus provide a new therapeutic approach to control tumor progression and bone destruction in patients with MM.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tomoya Takeda
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Kenji Mashimo
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan.,Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Yu-Ichi Koumoto
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Sachi Hoshida
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tatsuki Itoh
- Faculty of Agriculture, Department of Food Science and Nutrition, Kindai University, Nara, Nara, Japan
| | - Motohiro Imano
- Faculty of Medicine, Department of Surgery, Kindai University, Osakasayama, Osaka, Japan
| | - Takao Satou
- Faculty of Medicine, Department of Pathology, Kindai University, Osakasayama, Osaka, Japan
| | - Katsuhiko Sakaguchi
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Shozo Nishida
- Faculty of Pharmacy, Division of Pharmacotherapy, Kindai University, Kowakae, Higashi-Osaka, Japan
| |
Collapse
|
19
|
Qattan MY, Bakker EY, Rajendran R, Chen DWC, Saha V, Liu J, Zeef L, Schwartz JM, Mutti L, Demonacos C, Krstic-Demonacos M. Differential regulation of cell death pathways by the microenvironment correlates with chemoresistance and survival in leukaemia. PLoS One 2017; 12:e0178606. [PMID: 28582465 PMCID: PMC5459454 DOI: 10.1371/journal.pone.0178606] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/16/2017] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoids (GCs) and topoisomerase II inhibitors are used to treat acute lymphoblastic leukaemia (ALL) as they induce death in lymphoid cells through the glucocorticoid receptor (GR) and p53 respectively. Mechanisms underlying ALL cell death and the contribution of the bone marrow microenvironment to drug response/resistance remain unclear. The role of the microenvironment and the identification of chemoresistance determinants were studied by transcriptomic analysis in ALL cells treated with Dexamethasone (Dex), and Etoposide (Etop) grown in the presence or absence of bone marrow conditioned media (CM). The necroptotic (RIPK1) and the apoptotic (caspase-8/3) markers were downregulated by CM, whereas the inhibitory effects of chemotherapy on the autophagy marker Beclin-1 (BECN1) were reduced suggesting CM exerts cytoprotective effects. GCs upregulated the RIPK1 ubiquitinating factor BIRC3 (cIAP2), in GC-sensitive (CEM-C7-14) but not in resistant (CEM-C1-15) cells. In addition, CM selectively affected GR phosphorylation in a site and cell-specific manner. GR is recruited to RIPK1, BECN1 and BIRC3 promoters in the sensitive but not in the resistant cells with phosphorylated GR forms being generally less recruited in the presence of hormone. FACS analysis and caspase-8 assays demonstrated that CM promoted a pro-survival trend. High molecular weight proteins reacting with the RIPK1 antibody were modified upon incubation with the BIRC3 inhibitor AT406 in CEM-C7-14 cells suggesting that they represent ubiquitinated forms of RIPK1. Our data suggest that there is a correlation between microenvironment-induced ALL proliferation and altered response to chemotherapy.
Collapse
Affiliation(s)
- Malak Yahia Qattan
- College of Applied Medical Sciences and Community Services (CAMS&CS), King Saud University, Riyadh, Saudi Arabia
| | - Emyr Yosef Bakker
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Ramkumar Rajendran
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Daphne Wei-Chen Chen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Vaskar Saha
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Tata Translational Cancer Research Centre, Kolkata, India
| | - Jizhong Liu
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Leo Zeef
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jean-Marc Schwartz
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Luciano Mutti
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Constantinos Demonacos
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Marija Krstic-Demonacos
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
- * E-mail:
| |
Collapse
|
20
|
Martin JH, Bromfield EG, Aitken RJ, Nixon B. Biochemical alterations in the oocyte in support of early embryonic development. Cell Mol Life Sci 2017; 74:469-485. [PMID: 27604868 PMCID: PMC11107538 DOI: 10.1007/s00018-016-2356-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/28/2016] [Accepted: 09/01/2016] [Indexed: 01/01/2023]
Abstract
Notwithstanding the enormous reproductive potential encapsulated within a mature mammalian oocyte, these cells present only a limited window for fertilization before defaulting to an apoptotic cascade known as post-ovulatory oocyte aging. The only cell with the capacity to rescue this potential is the fertilizing spermatozoon. Indeed, the union of these cells sets in train a remarkable series of events that endows the oocyte with the capacity to divide and differentiate into the trillions of cells that comprise a new individual. Traditional paradigms hold that, beyond the initial stimulation of fluctuating calcium (Ca2+) required for oocyte activation, the fertilizing spermatozoon plays limited additional roles in the early embryo. While this model has now been drawn into question in view of the recent discovery that spermatozoa deliver developmentally important classes of small noncoding RNAs and other epigenetic modulators to oocytes during fertilization, it is nevertheless apparent that the primary responsibility for oocyte activation rests with a modest store of maternally derived proteins and mRNA accumulated during oogenesis. It is, therefore, not surprising that widespread post-translational modifications, in particular phosphorylation, hold a central role in endowing these proteins with sufficient functional diversity to initiate embryonic development. Indeed, proteins targeted for such modifications have been linked to oocyte activation, recruitment of maternal mRNAs, DNA repair and resumption of the cell cycle. This review, therefore, seeks to explore the intimate relationship between Ca2+ release and the suite of molecular modifications that sweep through the oocyte to ensure the successful union of the parental germlines and ensure embryogenic fidelity.
Collapse
Affiliation(s)
- Jacinta H Martin
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.
| | - Elizabeth G Bromfield
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - R John Aitken
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Brett Nixon
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| |
Collapse
|
21
|
Li Y, Buijs-Gladdines JGCAM, Canté-Barrett K, Stubbs AP, Vroegindeweij EM, Smits WK, van Marion R, Dinjens WNM, Horstmann M, Kuiper RP, Buijsman RC, Zaman GJR, van der Spek PJ, Pieters R, Meijerink JPP. IL-7 Receptor Mutations and Steroid Resistance in Pediatric T cell Acute Lymphoblastic Leukemia: A Genome Sequencing Study. PLoS Med 2016; 13:e1002200. [PMID: 27997540 PMCID: PMC5172551 DOI: 10.1371/journal.pmed.1002200] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Pediatric acute lymphoblastic leukemia (ALL) is the most common childhood cancer and the leading cause of cancer-related mortality in children. T cell ALL (T-ALL) represents about 15% of pediatric ALL cases and is considered a high-risk disease. T-ALL is often associated with resistance to treatment, including steroids, which are currently the cornerstone for treating ALL; moreover, initial steroid response strongly predicts survival and cure. However, the cellular mechanisms underlying steroid resistance in T-ALL patients are poorly understood. In this study, we combined various genomic datasets in order to identify candidate genetic mechanisms underlying steroid resistance in children undergoing T-ALL treatment. METHODS AND FINDINGS We performed whole genome sequencing on paired pre-treatment (diagnostic) and post-treatment (remission) samples from 13 patients, and targeted exome sequencing of pre-treatment samples from 69 additional T-ALL patients. We then integrated mutation data with copy number data for 151 mutated genes, and this integrated dataset was tested for associations of mutations with clinical outcomes and in vitro drug response. Our analysis revealed that mutations in JAK1 and KRAS, two genes encoding components of the interleukin 7 receptor (IL7R) signaling pathway, were associated with steroid resistance and poor outcome. We then sequenced JAK1, KRAS, and other genes in this pathway, including IL7R, JAK3, NF1, NRAS, and AKT, in these 69 T-ALL patients and a further 77 T-ALL patients. We identified mutations in 32% (47/146) of patients, the majority of whom had a specific T-ALL subtype (early thymic progenitor ALL or TLX). Based on the outcomes of these patients and their prednisolone responsiveness measured in vitro, we then confirmed that these mutations were associated with both steroid resistance and poor outcome. To explore how these mutations in IL7R signaling pathway genes cause steroid resistance and subsequent poor outcome, we expressed wild-type and mutant IL7R signaling molecules in two steroid-sensitive T-ALL cell lines (SUPT1 and P12 Ichikawa cells) using inducible lentiviral expression constructs. We found that expressing mutant IL7R, JAK1, or NRAS, or wild-type NRAS or AKT, specifically induced steroid resistance without affecting sensitivity to vincristine or L-asparaginase. In contrast, wild-type IL7R, JAK1, and JAK3, as well as mutant JAK3 and mutant AKT, had no effect. We then performed a functional study to examine the mechanisms underlying steroid resistance and found that, rather than changing the steroid receptor's ability to activate downstream targets, steroid resistance was associated with strong activation of MEK-ERK and AKT, downstream components of the IL7R signaling pathway, thereby inducing a robust antiapoptotic response by upregulating MCL1 and BCLXL expression. Both the MEK-ERK and AKT pathways also inactivate BIM, an essential molecule for steroid-induced cell death, and inhibit GSK3B, an important regulator of proapoptotic BIM. Importantly, treating our cell lines with IL7R signaling inhibitors restored steroid sensitivity. To address clinical relevance, we treated primary T-ALL cells obtained from 11 patients with steroids either alone or in combination with IL7R signaling inhibitors; we found that including a MEK, AKT, mTOR, or dual PI3K/mTOR inhibitor strongly increased steroid-induced cell death. Therefore, combining these inhibitors with steroid treatment may enhance steroid sensitivity in patients with ALL. The main limitation of our study was the modest cohort size, owing to the very low incidence of T-ALL. CONCLUSIONS Using an unbiased sequencing approach, we found that specific mutations in IL7R signaling molecules underlie steroid resistance in T-ALL. Future prospective clinical studies should test the ability of inhibitors of MEK, AKT, mTOR, or PI3K/mTOR to restore or enhance steroid sensitivity and improve clinical outcome.
Collapse
Affiliation(s)
- Yunlei Li
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Jessica G. C. A. M. Buijs-Gladdines
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kirsten Canté-Barrett
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Andrew P. Stubbs
- Department of Bioinformatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eric M. Vroegindeweij
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Willem K. Smits
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ronald van Marion
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Martin Horstmann
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Co-operative Study Group for Childhood Acute Lymphoblastic Leukemia, Hamburg, Germany
| | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | | - Rob Pieters
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jules P. P. Meijerink
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center/Sophia Children’s Hospital, Rotterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Froment R, Bédard C. Marked hyperphosphatasemia associated with an acute leukemia in a Great Dane. Vet Clin Pathol 2016; 45:459-65. [PMID: 27538028 DOI: 10.1111/vcp.12389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This is the report of a 5-year-old male neutered Great Dane with an extreme leukocytosis (544.9 × 10(9) cells/L; RI 5.2-13.9 × 10(9) cells/L) characterized by highly atypical round cells. Cellular morphologic features such as cytoplasmic membrane blebs, a high nuclear-to-cytoplasmic ratio, and nuclear indentations and irregularities and large nucleoli, as well as immunocytochemistry for CD3 and CD79, myeloperoxidase cytochemistry, and clonality testing were not conclusive for myeloid or lymphoid origin. Marked alkaline hyperphosphatasemia was present at the first visit (2783.0 U/L; RI 6-80.0 U/L), followed by a 5-fold increase (14,000 U/L) a week later, identified as being mostly contributed by the bone-ALP isoform (11,062 U/L; RI 0-30 U/L). In addition, the atypical leukocytes were strongly positive for cytoplasmic ALP activity. In vitro lysis of a heparin blood sample resulted in a 1.7-fold increase of ALP activity, supporting the origin of the hyperphosphatasemia at least in part from the leukemic cell population. To the authors' knowledge, this is a unique case of alkaline hyperphosphatasemia, due at least to a leukemic cell population producing a bone-ALP isoform, regardless of the exact nature of the leukemia.
Collapse
Affiliation(s)
- Rémi Froment
- Department of Pathology and Microbiology, Faculté de Médecine Vétérinaire, Université de Montréal, Saint Hyacinth, QC, Canada.
| | - Christian Bédard
- Department of Pathology and Microbiology, Faculté de Médecine Vétérinaire, Université de Montréal, Saint Hyacinth, QC, Canada
| |
Collapse
|
23
|
Polak A, Kiliszek P, Sewastianik T, Szydłowski M, Jabłońska E, Białopiotrowicz E, Górniak P, Markowicz S, Nowak E, Grygorowicz MA, Prochorec-Sobieszek M, Nowis D, Gołąb J, Giebel S, Lech-Marańda E, Warzocha K, Juszczyński P. MEK Inhibition Sensitizes Precursor B-Cell Acute Lymphoblastic Leukemia (B-ALL) Cells to Dexamethasone through Modulation of mTOR Activity and Stimulation of Autophagy. PLoS One 2016; 11:e0155893. [PMID: 27196001 PMCID: PMC4872998 DOI: 10.1371/journal.pone.0155893] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/05/2016] [Indexed: 01/16/2023] Open
Abstract
Resistance to glucocorticosteroids (GCs) is a major adverse prognostic factor in B-ALL, but the molecular mechanisms leading to GC resistance are not completely understood. Herein, we sought to elucidate the molecular background of GC resistance in B-ALL and characterize the therapeutic potential of targeted intervention in these mechanisms. Using exploratory bioinformatic approaches, we found that resistant cells exhibited significantly higher expression of MEK/ERK (MAPK) pathway components. We found that GC-resistant ALL cell lines had markedly higher baseline activity of MEK and small-molecule MEK1/2 inhibitor selumetinib increased GCs-induced cell death. MEK inhibitor similarly increased in vitro dexamethasone activity in primary ALL blasts from 19 of 22 tested patients. To further confirm these observations, we overexpressed a constitutively active MEK mutant in GC-sensitive cells and found that forced MEK activity induced resistance to dexamethasone. Since recent studies highlight the role GC-induced autophagy upstream of apoptotic cell death, we assessed LC3 processing, MDC staining and GFP-LC3 relocalization in cells incubated with either DEX, SEL or combination of drugs. Unlike either drug alone, only their combination markedly increased these markers of autophagy. These changes were associated with decreased mTOR activity and blocked 4E-BP1 phosphorylation. In cells with silenced beclin-1 (BCN1), required for autophagosome formation, the synergy of DEX and SEL was markedly reduced. Taken together, we show that MEK inhibitor selumetinib enhances dexamethasone toxicity in GC-resistant B-ALL cells. The underlying mechanism of this interaction involves inhibition of mTOR signaling pathway and modulation of autophagy markers, likely reflecting induction of this process and required for cell death. Thus, our data demonstrate that modulation of MEK/ERK pathway is an attractive therapeutic strategy overcoming GC resistance in B-ALL patients.
Collapse
Affiliation(s)
- Anna Polak
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Przemysław Kiliszek
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Tomasz Sewastianik
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Maciej Szydłowski
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Ewa Jabłońska
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Emilia Białopiotrowicz
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Patryk Górniak
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Dept. of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Sergiusz Markowicz
- Dept. of Immunology, Maria Sklodowska-Curie Memorial Cancer Center–Institute of Oncology, Warsaw, Poland
| | - Eliza Nowak
- Dept. of Immunology, Maria Sklodowska-Curie Memorial Cancer Center–Institute of Oncology, Warsaw, Poland
| | - Monika A. Grygorowicz
- Dept. of Immunology, Maria Sklodowska-Curie Memorial Cancer Center–Institute of Oncology, Warsaw, Poland
| | | | - Dominika Nowis
- Genomic Medicine, Dept. of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Jakub Gołąb
- Dept. of Immunology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Sebastian Giebel
- Dept. of Bone Marrow Transplantation and Hematology-Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Ewa Lech-Marańda
- Dept. of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Dept. of Hematology and Transfusion Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Krzysztof Warzocha
- Dept. of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Przemysław Juszczyński
- Dept. of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- * E-mail:
| |
Collapse
|
24
|
Sionov RV, Vlahopoulos SA, Granot Z. Regulation of Bim in Health and Disease. Oncotarget 2015; 6:23058-134. [PMID: 26405162 PMCID: PMC4695108 DOI: 10.18632/oncotarget.5492] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/08/2015] [Indexed: 11/25/2022] Open
Abstract
The BH3-only Bim protein is a major determinant for initiating the intrinsic apoptotic pathway under both physiological and pathophysiological conditions. Tight regulation of its expression and activity at the transcriptional, translational and post-translational levels together with the induction of alternatively spliced isoforms with different pro-apoptotic potential, ensure timely activation of Bim. Under physiological conditions, Bim is essential for shaping immune responses where its absence promotes autoimmunity, while too early Bim induction eliminates cytotoxic T cells prematurely, resulting in chronic inflammation and tumor progression. Enhanced Bim induction in neurons causes neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Moreover, type I diabetes is promoted by genetically predisposed elevation of Bim in β-cells. On the contrary, cancer cells have developed mechanisms that suppress Bim expression necessary for tumor progression and metastasis. This review focuses on the intricate network regulating Bim activity and its involvement in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| | - Spiros A. Vlahopoulos
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Thivon and Levadias, Goudi, Athens, Greece
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
25
|
Mechanisms of Drug Resistance in Veterinary Oncology- A Review with an Emphasis on Canine Lymphoma. Vet Sci 2015; 2:150-184. [PMID: 29061939 PMCID: PMC5644636 DOI: 10.3390/vetsci2030150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/22/2015] [Accepted: 08/04/2015] [Indexed: 01/09/2023] Open
Abstract
Drug resistance (DR) is the major limiting factor in the successful treatment of systemic neoplasia with cytotoxic chemotherapy. DR can be either intrinsic or acquired, and although the development and clinical implications are different, the underlying mechanisms are likely to be similar. Most causes for DR are pharmacodynamic in nature, result from adaptations within the tumor cell and include reduced drug uptake, increased drug efflux, changes in drug metabolism or drug target, increased capacity to repair drug-induced DNA damage or increased resistance to apoptosis. The role of active drug efflux transporters, and those of the ABC-transporter family in particular, have been studied extensively in human oncology and to a lesser extent in veterinary medicine. Methods reported to assess ABC-transporter status include detection of the actual protein (Western blot, immunohistochemistry), mRNA or ABC-transporter function. The three major ABC-transporters associated with DR in human oncology are ABCB1 or P-gp, ABCC1 or MRP1, and ABCG2 or BCRP, and have been demonstrated in canine cell lines, healthy dogs and dogs with cancer. Although this supports a causative role for these ABC-transporters in DR cytotoxic agents in the dog, the relative contribution to the clinical phenotype of DR in canine cancer remains an area of debate and requires further prospective studies.
Collapse
|
26
|
Goodwin JE. Glucocorticoids and the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [DOI: 10.1007/978-1-4939-2895-8_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
27
|
Palagani A, Op de Beeck K, Naulaerts S, Diddens J, Sekhar Chirumamilla C, Van Camp G, Laukens K, Heyninck K, Gerlo S, Mestdagh P, Vandesompele J, Berghe WV. Ectopic microRNA-150-5p transcription sensitizes glucocorticoid therapy response in MM1S multiple myeloma cells but fails to overcome hormone therapy resistance in MM1R cells. PLoS One 2014; 9:e113842. [PMID: 25474406 PMCID: PMC4256227 DOI: 10.1371/journal.pone.0113842] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 11/01/2014] [Indexed: 11/18/2022] Open
Abstract
Glucocorticoids (GCs) selectively trigger cell death in the multiple myeloma cell line MM1S which express NR3C1/Glucocorticoid Receptor (GR) protein, but fail to kill MM1R cells which lack GR protein. Given recent demonstrations of altered microRNA profiles in a diverse range of haematological malignancies and drug resistance, we characterized GC inducible mRNA and microRNA transcription profiles in GC sensitive MM1S as compared to GC resistant MM1R cells. Transcriptome analysis revealed that GCs regulate expression of multiple genes involved in cell cycle control, cell organization, cell death and immunological disease in MM1S cells, which remain unaffected in MM1R cells. With respect to microRNAs, mir-150-5p was identified as the most time persistent GC regulated microRNA, out of 5 QPCR validated microRNAs (mir-26b, mir-125a-5p, mir-146-5p, mir-150-5p, and mir-184), which are GC inducible in MM1S but not in MM1R cells. Functional studies further revealed that ectopic transfection of a synthetic mir-150-5p mimics GR dependent gene expression changes involved in cell death and cell proliferation pathways. Remarkably, despite the gene expression changes observed, overexpression of mir-150-5p in absence of GCs did not trigger significant cytotoxicity in MM1S or MM1R cells. This suggests the requirement of additional steps in GC induced cell death, which can not be mimicked by mir-150-5p overexpression alone. Interestingly, a combination of mir-150-5p transfection with low doses GC in MM1S cells was found to sensitize therapy response, whereas opposite effects could be observed with a mir-150-5p specific antagomir. Although mir-150-5p overexpression did not substantially change GR expression levels, it was found that mir-150-5p evokes GR specific effects through indirect mRNA regulation of GR interacting transcription factors and hormone receptors, GR chaperones, as well as various effectors of unfolded protein stress and chemokine signalling. Altogether GC-inducible mir-150-5p adds another level of regulation to GC specific therapeutic responses in multiple myeloma.
Collapse
Affiliation(s)
- Ajay Palagani
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Physiology, Ghent University, Ghent, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Cancer Research and Clinical Oncology, Department of Medical Oncology, University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
| | - Stefan Naulaerts
- Biomedical Informatics Research Center Antwerp (Biomina), University of Antwerp & University Hospital Antwerp, Antwerp, Belgium
- Advanced Database Research and Modelling (ADReM), Department of Mathematics & Computer sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Jolien Diddens
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Chandra Sekhar Chirumamilla
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Kris Laukens
- Biomedical Informatics Research Center Antwerp (Biomina), University of Antwerp & University Hospital Antwerp, Antwerp, Belgium
- Advanced Database Research and Modelling (ADReM), Department of Mathematics & Computer sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Karen Heyninck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Physiology, Ghent University, Ghent, Belgium
| | - Sarah Gerlo
- VIB-UGent Department of Medical Protein Research, Ghent, Belgium
| | - Pieter Mestdagh
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Joke Vandesompele
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Physiology, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
28
|
Orlovsky M, Dosenko V, Spiga F, Skibo G, Lightman S. Hippocampus remodeling by chronic stress accompanied by GR, proteasome and caspase-3 overexpression. Brain Res 2014; 1593:83-94. [DOI: 10.1016/j.brainres.2014.09.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 12/31/2022]
|
29
|
|
30
|
Fan Z, Sehm T, Rauh M, Buchfelder M, Eyupoglu IY, Savaskan NE. Dexamethasone alleviates tumor-associated brain damage and angiogenesis. PLoS One 2014; 9:e93264. [PMID: 24714627 PMCID: PMC3979667 DOI: 10.1371/journal.pone.0093264] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 03/03/2014] [Indexed: 01/05/2023] Open
Abstract
Children and adults with the most aggressive form of brain cancer, malignant gliomas or glioblastoma, often develop cerebral edema as a life-threatening complication. This complication is routinely treated with dexamethasone (DEXA), a steroidal anti-inflammatory drug with pleiotropic action profile. Here we show that dexamethasone reduces murine and rodent glioma tumor growth in a concentration-dependent manner. Low concentrations of DEXA are already capable of inhibiting glioma cell proliferation and at higher levels induce cell death. Further, the expression of the glutamate antiporter xCT (system Xc−; SLC7a11) and VEGFA is up-regulated after DEXA treatment indicating early cellular stress responses. However, in human gliomas DEXA exerts differential cytotoxic effects, with some human glioma cells (U251, T98G) resistant to DEXA, a finding corroborated by clinical data of dexamethasone non-responders. Moreover, DEXA-resistant gliomas did not show any xCT alterations, indicating that these gene expressions are associated with DEXA-induced cellular stress. Hence, siRNA-mediated xCT knockdown in glioma cells increased the susceptibility to DEXA. Interestingly, cell viability of primary human astrocytes and primary rodent neurons is not affected by DEXA. We further tested the pharmacological effects of DEXA on brain tissue and showed that DEXA reduces tumor-induced disturbances of the microenvironment such as neuronal cell death and tumor-induced angiogenesis. In conclusion, we demonstrate that DEXA inhibits glioma cell growth in a concentration and species-dependent manner. Further, DEXA executes neuroprotective effects in brains and reduces tumor-induced angiogenesis. Thus, our investigations reveal that DEXA acts pleiotropically and impacts tumor growth, tumor vasculature and tumor-associated brain damage.
Collapse
Affiliation(s)
- Zheng Fan
- Department of Neurosurgery, University Hospital of Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
| | - Tina Sehm
- Department of Neurosurgery, University Hospital of Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University Hospital of Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
| | - Ilker Y. Eyupoglu
- Department of Neurosurgery, University Hospital of Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
| | - Nicolai E. Savaskan
- Department of Neurosurgery, University Hospital of Erlangen, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Bavaria, Germany
- * E-mail:
| |
Collapse
|
31
|
EMP1, a novel poor prognostic factor in pediatric leukemia regulates prednisolone resistance, cell proliferation, migration and adhesion. Leukemia 2014; 28:1828-37. [PMID: 24625531 DOI: 10.1038/leu.2014.80] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/03/2014] [Accepted: 02/12/2014] [Indexed: 01/21/2023]
Abstract
Still 20% of pediatric acute lymphoblastic leukemia (ALL) patients relapse on or after current treatment strategies. Treatment failure is associated with resistance to prednisolone. We aimed to find new druggable targets that modulate prednisolone resistance. We generated microarray gene expression profiles of 256 pediatric ALL patient samples and identified a 3.4-fold increase in epithelial membrane protein 1 (EMP1) expression in in vitro prednisolone-resistant compared with -sensitive patients (P=0.003). EMP1 silencing in six precursor-B ALL (BCP-ALL) and T-ALL cell lines induced apoptosis and cell-cycle arrest leading to 84.1±4.5% reduction in survival compared with non-silencing control transduced cells (non-silencing control short hairpin, shNSC) (P=0.014). Moreover, EMP1 silencing sensitized to prednisolone up to 18.8-fold (P<0.001). EMP1 silencing also abrogated migration and adhesion to mesenchymal stromal cells (MSCs) by 78.3±9.0 and 29.3±4.1% compared with shNSC (P<0.05). We discovered that EMP1 contributes to MSC-mediated prednisolone resistance. Pathway analysis indicated that EMP1 signals through the Src kinase family. EMP1-high BCP-ALL patients showed a poorer 5-year event-free survival compared with EMP1-low patients (77±2 vs. 89±2%, P=0.003). Multivariate analysis taking along white blood cell count, age, prednisolone resistance and subtype identified EMP1 as an independent predictor for poor outcome in BCP-ALL (P=0.004, hazard ratio: 2.36 (1.31-4.25). This study provides preclinical evidence that EMP1 is an interesting candidate for drug development to optimize treatment of BCP-ALL.
Collapse
|
32
|
Kfir-Erenfeld S, Yefenof E. Non-genomic events determining the sensitivity of hemopoietic malignancies to glucocorticoid-induced apoptosis. Cancer Immunol Immunother 2014; 63:37-43. [PMID: 24072402 PMCID: PMC11028523 DOI: 10.1007/s00262-013-1477-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 09/13/2013] [Indexed: 01/22/2023]
Abstract
Glucocorticoid (GC) hormones have been introduced as therapeutic agents in blood cancers six decades ago. The effectiveness of GC treatment stems from its ability to induce apoptotic death of hemopoietic cells. A major impediment in GC therapy is the acquisition of resistance to the drug upon repeated treatment. In addition, some blood cancers are a priori resistant to GC therapy. Usually, resistance to GC correlates with poor prognosis. Albeit the wide use of GC in clinical practice, their mode of action is not fully understood. The cellular response to GC is initiated by its binding to the cytosolic GC receptor (GR) that translocates to the nucleus and modulates gene expression. However, nuclear activities of GR occur in both apoptosis-sensitive and apoptosis-resistant cells. These apparent controversies can be resolved by deciphering non-genomic effects of GCs and the mode by which they modulate the apoptotic response. We suggest that non-genomic consequences of GC stimulation determine the cell fate toward survival or death. Understanding the cellular mechanisms of GC apoptotic sensitivity contributes to the development of new modalities for overcoming GC resistance.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, POB: 12272, 91120, Jerusalem, Israel,
| | | |
Collapse
|
33
|
Microfluidic chip integrated with flexible PDMS-based electrochemical cytosensor for dynamic analysis of drug-induced apoptosis on HeLa cells. Biosens Bioelectron 2014; 51:97-102. [DOI: 10.1016/j.bios.2013.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/06/2013] [Accepted: 07/12/2013] [Indexed: 12/26/2022]
|
34
|
Zandvliet M, Rutteman G, Teske E. Prednisolone inclusion in a first-line multidrug cytostatic protocol for the treatment of canine lymphoma does not affect therapy results. Vet J 2013; 197:656-61. [DOI: 10.1016/j.tvjl.2013.04.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/25/2013] [Accepted: 04/29/2013] [Indexed: 11/26/2022]
|
35
|
Tung S, Shi Y, Wong K, Zhu F, Gorczynski R, Laister RC, Minden M, Blechert AK, Genzel Y, Reichl U, Spaner DE. PPARα and fatty acid oxidation mediate glucocorticoid resistance in chronic lymphocytic leukemia. Blood 2013; 122:969-80. [PMID: 23814018 DOI: 10.1182/blood-2013-03-489468] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
High-dose glucocorticoids (GCs) can be a useful treatment for aggressive forms of chronic lymphocytic leukemia (CLL). However, their mechanism of action is not well understood, and resistance to GCs is inevitable. In a minimal, serum-free culture system, the synthetic GC dexamethasone (DEX) was found to decrease the metabolic activity of CLL cells, indicated by down-regulation of pyruvate kinase M2 (PKM2) expression and activity, decreased levels of pyruvate and its metabolites, and loss of mitochondrial membrane potential. This metabolic restriction was associated with decreased size and death of some of the tumor cells in the population. Concomitant plasma membrane damage increased killing of CLL cells by DEX. However, the nuclear receptor peroxisome proliferator activated receptor α (PPARα), which regulates fatty acid oxidation, was also increased by DEX, and adipocyte-derived lipids, lipoproteins, and propionic acid protected CLL cells from DEX. PPARα and fatty acid oxidation enzyme inhibitors increased DEX-mediated killing of CLL cells in vitro and clearance of CLL xenografts in vivo. These findings suggest that GCs prevent tumor cells from generating the energy needed to repair membrane damage, fatty acid oxidation is a mechanism of resistance to GC-mediated cytotoxicity, and PPARα inhibition is a strategy to improve the therapeutic efficacy of GCs.
Collapse
MESH Headings
- Adipocytes/cytology
- Animals
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Membrane/metabolism
- Culture Media, Conditioned
- Dexamethasone/pharmacology
- Drug Resistance, Neoplasm
- Fatty Acids/metabolism
- Gene Expression Regulation, Leukemic
- Glucocorticoids/pharmacology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lipid Metabolism
- Membrane Potential, Mitochondrial
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Oxygen/metabolism
- PPAR alpha/metabolism
- Phosphorylation
- Propionates/chemistry
- Thyroid Hormones/metabolism
- Thyroid Hormone-Binding Proteins
Collapse
Affiliation(s)
- Stephanie Tung
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute, 2075 Bayview Ave., Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bhatt AJ, Feng Y, Wang J, Famuyide M, Hersey K. Dexamethasone induces apoptosis of progenitor cells in the subventricular zone and dentate gyrus of developing rat brain. J Neurosci Res 2013; 91:1191-202. [PMID: 23686666 DOI: 10.1002/jnr.23232] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/09/2013] [Accepted: 03/12/2013] [Indexed: 01/22/2023]
Abstract
The use of dexamethasone in premature infants to prevent and/or treat bronchopulmonary dysplasia adversely affects neurocognitive development and is associated with cerebral palsy. The underlying mechanisms of these effects are multifactorial and likely include apoptosis. The objective of this study was to confirm whether dexamethasone causes apoptosis in different regions of the developing rat brain. On postnatal day 2, pups in each litter were randomly divided into the dexamethasone-treated (n = 91) or vehicle-treated (n = 92) groups. Rat pups in the dexamethasone group received tapering doses of dexamethasone on postnatal days 3-6 (0.5, 0.25, 0.125, and 0.06 mg/kg/day, respectively). Dexamethasone treatment significantly decreased the gain of body and brain weight and increased brain caspase-3 activity, DNA fragments, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, and cleaved caspse-3-positive cells at 24 hr after treatment. Dexamethasone increased cleaved caspse-3-positive cells in the cortex, thalamus, hippocampus, cerebellum, dentate gyrus, and subventricular zone. Double-immunofluorescence studies show that progenitor cells in the subventricular zone and dentate gyrus preferentially undergo apoptosis following dexamethasone exposure. These results indicate that dexamethasone-induced apoptosis in immature cells in developing brain is one of the mechanisms of its neurodegenerative effects in newborn rats.
Collapse
Affiliation(s)
- Abhay J Bhatt
- Division of Newborn Medicine, Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| | | | | | | | | |
Collapse
|
37
|
Lee SR, Kim HK, Song IS, Youm J, Dizon LA, Jeong SH, Ko TH, Heo HJ, Ko KS, Rhee BD, Kim N, Han J. Glucocorticoids and their receptors: insights into specific roles in mitochondria. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 112:44-54. [PMID: 23603102 DOI: 10.1016/j.pbiomolbio.2013.04.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/27/2022]
Abstract
Glucocorticoids (GCs) affect most physiological systems and are the most frequently used drugs for multiple disorders and organ transplantation. GC functions depend on a balance between circulating GC and cytoplasmic glucocorticoid receptor II (GR). Mitochondria individually enclose circular, double-stranded DNA that is expressed and replicated in response to nuclear-encoded factors imported from the cytoplasm. Fine-tuning and response to cellular demands should be coordinately regulated by the nucleus and mitochondria; thus mitochondrial-nuclear interaction is vital to optimal mitochondrial function. Elucidation of the direct and indirect effects of steroids, including GCs, on mitochondria is an important and emerging field of research. Mitochondria may also be under GC control because GRs are present in mitochondria, and glucocorticoid response elements (GREs) reside in the mitochondrial genome. Therefore, mitochondrial gene expression can be regulated by GCs via at least two different mechanisms: direct action on mitochondrial DNA and oxidative phosphorylation (OXPHOS) genes, or by an indirect effect through interaction with nuclear genes. In this review, we outline possible mechanisms of regulation of mitochondrial genes in response to GCs in view of translocation of the GR into mitochondria and the possible regulation of OXPHOS genes by GREs in the mitochondrial genome.
Collapse
Affiliation(s)
- Sung-Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, 633-165 Gaegeum-Dong, Busanjin-Gu, 613-735 Busan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gonzalez OA, Novak MJ, Kirakodu S, Stromberg AJ, Shen S, Orraca L, Gonzalez-Martinez J, Ebersole JL. Effects of aging on apoptosis gene expression in oral mucosal tissues. Apoptosis 2013; 18:249-59. [PMID: 23334583 PMCID: PMC3592930 DOI: 10.1007/s10495-013-0806-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Apoptotic processes are important for physiologic renewal of an intact epithelial barrier and contribute some antimicrobial resistance for bacteria and viruses, as well as anti-inflammatory effects that benefits the mucosa. The oral cavity presents a model of host-bacterial interactions at mucosal surfaces, in which a panoply of microorganisms colonizes various niches in the oral cavity and creates complex multispecies biofilms that challenge the gingival tissues. This report details gene expression in apoptotic pathways that occur in oral mucosal tissues across the lifespan, using a nonhuman primate model. Macaca mulatta primates from 2 to 23 years of age (n = 23) were used in a cross-sectional study to obtain clinical healthy gingival tissues specimens. Further, mRNA was prepared and evaluated using the Affymetrix Rhesus GeneChip and 88 apoptotic pathway genes were evaluated. The results identified significant positive correlations with age in 12 genes and negative correlations with an additional five genes. The gene effects were predicted to alter apoptosis receptor levels, extrinsic apoptotic pathways through caspases, cytokine effects on apoptotic events, Ca(+2)-induced death signaling, cell cycle checkpoints, and potential effects of survival factors. Both the positively and negatively correlated genes within the apoptotic pathways provided evidence that healthy tissues in aging animals exhibit decreased apoptotic potential compared to younger animals. The results suggested that decreased physiologic apoptotic process in the dynamic septic environment of the oral mucosal tissues could increase the risk of aging tissues to undergo destructive disease processes through dysregulated inflammatory responses to the oral microbial burden.
Collapse
Affiliation(s)
- Octavio A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, 1095 VA Drive, HSRB 414, Lexington, KY 40536-0305, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Sionov RV. MicroRNAs and Glucocorticoid-Induced Apoptosis in Lymphoid Malignancies. ISRN HEMATOLOGY 2013; 2013:348212. [PMID: 23431463 PMCID: PMC3569899 DOI: 10.1155/2013/348212] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 11/14/2012] [Indexed: 12/20/2022]
Abstract
The initial response of lymphoid malignancies to glucocorticoids (GCs) is a critical parameter predicting successful treatment. Although being known as a strong inducer of apoptosis in lymphoid cells for almost a century, the signaling pathways regulating the susceptibility of the cells to GCs are only partly revealed. There is still a need to develop clinical tests that can predict the outcome of GC therapy. In this paper, I discuss important parameters modulating the pro-apoptotic effects of GCs, with a specific emphasis on the microRNA world comprised of small players with big impacts. The journey through the multifaceted complexity of GC-induced apoptosis brings forth explanations for the differential treatment response and raises potential strategies for overcoming drug resistance.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Department of Biochemistry and Molecular Biology, The Institute for Medical Research-Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Ein-Kerem, 91120 Jerusalem, Israel
| |
Collapse
|
40
|
Lee SR, Kim HK, Youm JB, Dizon LA, Song IS, Jeong SH, Seo DY, Ko KS, Rhee BD, Kim N, Han J. Non-genomic effect of glucocorticoids on cardiovascular system. Pflugers Arch 2012; 464:549-59. [PMID: 23001133 DOI: 10.1007/s00424-012-1155-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 09/05/2012] [Indexed: 01/01/2023]
Abstract
Glucocorticoids (GCs) are essential steroid hormones for homeostasis, development, metabolism, and cognition and possess anti-inflammatory and immunosuppressive actions. Since glucocorticoid receptor II (GR) is nearly ubiquitous, chronic activation or depletion of GCs leads to dysfunction of diverse organs, including the heart and blood vessels, resulting predominantly from changes in gene expression. Most studies, therefore, have focused on the genomic effects of GC to understand its related pathophysiological manifestations. The nongenomic effects of GCs clearly differ from well-known genomic effects, with the former responding within several minutes without the need for protein synthesis. There is increasing evidence that the nongenomic actions of GCs influence various physiological functions. To develop a GC-mediated therapeutic target for the treatment of cardiovascular disease, understanding the genomic and nongenomic effects of GC on the cardiovascular system is needed. This article reviews our current understanding of the underlying mechanisms of GCs on cardiovascular diseases and stress, as well as how nongenomic GC signaling contributes to these conditions. We suggest that manipulation of GC action based on both GC and GR metabolism, mitochondrial impact, and the action of serum- and glucocorticoid-dependent kinase 1 may provide new information with which to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Sung Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, 633-165 Gaegeum-Dong, Busanjin-Gu, Busan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zheng Y, Yang J, Qian J, Qiu P, Hanabuchi S, Lu Y, Wang Z, Liu Z, Li H, He J, Lin P, Weber D, Davis RE, Kwak L, Cai Z, Yi Q. PSGL-1/selectin and ICAM-1/CD18 interactions are involved in macrophage-induced drug resistance in myeloma. Leukemia 2012; 27:702-10. [PMID: 22996336 DOI: 10.1038/leu.2012.272] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemoresistance is the major obstacle in multiple myeloma (MM) management. We previously showed that macrophages protect myeloma cells, on a cell contact basis, from melphalan or dexamethasone-induced apoptosis in vitro. In this study, we found that macrophage-mediated myeloma drug resistance was also seen with purified macrophages from myeloma patients' bone marrow (BM) in vitro and was confirmed in vivo using the human myeloma-SCID (severe combined immunodeficient) mouse model. By profiling differentially regulated and paired plasma membrane protein genes, we showed that PSGL-1 (P-selectin glycoprotein ligand-1)/selectins and ICAM-1/CD18 played an important role in macrophage-mediated myeloma cell drug resistance, as blocking antibodies against these molecules or genetic knockdown of PSGL-1 or ICAM-1 in myeloma cells repressed macrophages' ability to protect myeloma cells. Interaction of macrophages and myeloma cells via these molecules activated Src and Erk1/2 kinases and c-myc pathways and suppressed caspase activation induced by chemotherapy drugs. Thus, our study sheds new light on the mechanism of drug resistance in MM and provides novel targets for improving the efficacy of chemotherapy in patients.
Collapse
Affiliation(s)
- Y Zheng
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rubio-Patiño C, Palmeri CM, Pérez-Perarnau A, Cosialls AM, Moncunill-Massaguer C, González-Gironès DM, Pons-Hernández L, López JM, Ventura F, Gil J, Pons G, Iglesias-Serret D. Glycogen synthase kinase-3β is involved in ligand-dependent activation of transcription and cellular localization of the glucocorticoid receptor. Mol Endocrinol 2012; 26:1508-20. [PMID: 22771494 DOI: 10.1210/me.2011-1366] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (GC) induce cell cycle arrest and apoptosis in different cell types and therefore are widely used to treat a variety of diseases including autoimmune disorders and cancer. This effect is mediated by the GC receptor (GR), a ligand-activated transcription factor that translocates into the nucleus where it modulates transcription of target genes in a promoter-specific manner. Glycogen synthase kinase-3 (GSK3) regulates GR response by genomic and nongenomic mechanisms, although the specific role of each isoform is not well defined. We used GSK3 pharmacological inhibitors and isoform-specific small interfering RNA to evaluate the role of GSK3 in the genomic regulation induced by GC. GSK3 inhibition resulted in the reduction of GC-induced mRNA expression of GC-induced genes such as BIM, HIAP1, and GILZ. Knockdown of GSK3β but not GSK3α reduced endogenous GILZ induction in response to dexamethasone and GR-dependent reporter gene activity. Chromatin immunoprecipitation experiments revealed that GSK3 inhibition impaired the dexamethasone-mediated binding of GR and RNA polymerase II to endogenous GILZ promoter. These results indicate that GSK3β is important for GR transactivation activity and that GSK3β inhibition suppresses GC-stimulated gene expression. Furthermore, we show that genomic regulation by the GR is independent of known GSK3β phosphorylation sites. We propose that GC-dependent transcriptional activation requires functional GSK3β signaling and that altered GSK3β activity influences cell response to GC.
Collapse
Affiliation(s)
- Camila Rubio-Patiño
- Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Reich E, Tamary A, Sionov RV, Melloul D. Involvement of thioredoxin-interacting protein (TXNIP) in glucocorticoid-mediated beta cell death. Diabetologia 2012; 55:1048-57. [PMID: 22246375 DOI: 10.1007/s00125-011-2422-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/07/2011] [Indexed: 11/29/2022]
Abstract
AIM/HYPOTHESIS Glucocorticoid hormones (GCs) are widely used to treat a variety of inflammatory and immune diseases. However, their long-term administration is associated with adverse metabolic effects, including glucose intolerance and diabetes. Our objective was to elucidate the mechanisms by which GCs affect beta cell survival with a specific emphasis on the role of the thioredoxin-interacting protein (TXNIP) in beta cell apoptosis. METHODS Human and mouse islets, together with MIN6 beta cells, were exposed to dexamethasone (Dex) and apoptosis was assessed by measuring the percentage of sub-G1 cells, the appearance of cleaved caspase-3 or by using a TUNEL assay. Dex-upregulated expression of Txnip mRNA was analysed by real-time PCR, and GC-modulated production and modification of proteins were determined by western blotting. RESULTS We provide evidence that TXNIP, a negative regulator of the antioxidant thioredoxin (TRX), is strongly induced in beta cells by GCs and that its induction is dependent on p38 mitogen-activated protein kinase (MAPK) activation. TXNIP downregulation by RNA interference, overexpression of the radical scavenger TRX1 or elevation of intracellular cAMP levels attenuated the Dex-mediated apoptosis. Dex-induced Txnip expression and beta cell apoptosis are mediated by the glucocorticoid receptor (GR), as the GR antagonist RU486 fully abolishes these effects. CONCLUSIONS/INTERPRETATION Altogether, our data suggest TXNIP as a novel mediator of GC-induced apoptosis in beta cells and further contribute to our understanding of beta cell death pathways.
Collapse
Affiliation(s)
- E Reich
- Department of Endocrinology, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | |
Collapse
|
44
|
Sifakis EG, Lambrou GI, Prentza A, Vlahopoulos S, Koutsouris D, Tzortzatou-Stathopoulou F, Chatziioannou AA. Elucidating the identity of resistance mechanisms to prednisolone exposure in acute lymphoblastic leukemia cells through transcriptomic analysis: A computational approach. J Clin Bioinforma 2011; 1:36. [PMID: 22185641 PMCID: PMC3313905 DOI: 10.1186/2043-9113-1-36] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 12/20/2011] [Indexed: 02/06/2023] Open
Abstract
Background It has been shown previously that glucocorticoids exert a dual mechanism of action, entailing cytotoxic, mitogenic as well as cell proliferative and anti-apoptotic responses, in a dose-dependent manner on CCRF-CEM cells at 72 h. Early gene expression response implies a dose-dependent dual mechanism of action of prednisolone too, something reflected on cell state upon 72 h of treatment. Methods In this work, a generic, computational microarray data analysis framework is proposed, in order to examine the hypothesis, whether CCRF-CEM cells exhibit an intrinsic or acquired mechanism of resistance and investigate the molecular imprint of this, upon prednisolone treatment. The experimental design enables the examination of both the dose (0 nM, 10 nM, 22 uM, 700 uM) effect of glucocorticoid exposure and the dynamics (early and late, namely 4 h, 72 h) of the molecular response of the cells at the transcriptomic layer. Results In this work, we demonstrated that CCRF-CEM cells may attain a mixed mechanism of response to glucocorticoids, however, with a clear preference towards an intrinsic mechanism of resistance. Specifically, at 4 h, prednisolone appeared to down-regulate apoptotic genes. Also, low and high prednisolone concentrations up-regulates genes related to metabolism and signal-transduction in both time points, thus favoring cell proliferative actions. In addition, regulation of NF-κB-related genes implies an inherent mechanism of resistance through the established link of NF-κB inflammatory role and GC-induced resistance. The analysis framework applied here highlights prednisolone-activated regulatory mechanisms through identification of early responding sets of genes. On the other hand, study of the prolonged exposure to glucocorticoids (72 h exposure) highlights the effect of homeostatic feedback mechanisms of the treated cells. Conclusions Overall, it appears that CCRF-CEM cells in this study exhibit a diversified, combined pattern of intrinsic and acquired resistance to prednisolone, with a tendency towards inherent resistant characteristics, through activation of different molecular courses of action.
Collapse
Affiliation(s)
- Emmanouil G Sifakis
- School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - George I Lambrou
- First Department of Pediatrics, University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Andriana Prentza
- Department of Digital Systems, University of Piraeus, Piraeus, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Dimitris Koutsouris
- School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | | | | |
Collapse
|
45
|
Liu Y, Chen LH, Yuan YW, Li QS, Sun AM, Guan J. Activation of AKT is associated with metastasis of nasopharyngeal carcinoma. Tumour Biol 2011; 33:241-5. [PMID: 22116667 DOI: 10.1007/s13277-011-0272-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 11/07/2011] [Indexed: 11/26/2022] Open
Abstract
Although radiotherapy results of nasopharyngeal carcinoma (NPC) at an early stage are better than other tumors, there is still a portion of patients with NPC who die before 5 years after the treatment; the underlying mechanism remains to be further understood. This study aims to investigate the mechanism by which NPC cells escape from irradiation. Patients with NPC at stage I was included in this study. All the patients were treated with irradiation. NPC biopsies were obtained from each patient before and 1 week after the start of radiotherapy. Expression of AKT in NPC tissue was assessed by Western blotting. NPC cell line, SUNE-1 cells, was treated with irradiation. The levels of AKT in SUNE-1 cells were also assessed. The frequency of apoptotic SUNE-1 cells was evaluated by flow cytometry. The levels of AKT were markedly increased in NPC tissue after treatment with irradiation, which was significantly correlated with NPC metastasis and mortality. After irradiation, NPC cell line, SUNE-1 cells, expressed higher levels of AKT than control cells. The knockdown of AKT in SUNE-1 cells markedly increased apoptotic cell rate. Radiotherapy can increase the levels of AKT in NPC cells that are associated with NPC metastasis and increase in mortality.
Collapse
Affiliation(s)
- Ying Liu
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | | | | | | | | | | |
Collapse
|
46
|
Zambon AC, Wilderman A, Ho A, Insel PA. Increased expression of the pro-apoptotic protein BIM, a mechanism for cAMP/protein kinase A (PKA)-induced apoptosis of immature T cells. J Biol Chem 2011; 286:33260-7. [PMID: 21808067 PMCID: PMC3190928 DOI: 10.1074/jbc.m111.268979] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 07/15/2011] [Indexed: 01/09/2023] Open
Abstract
The second messenger cAMP is proapoptotic for numerous cell types, but the mechanism for this proapoptotic action is not defined. Here, we use murine CD4(+)/CD8(+) S49 lymphoma cells and isolated thymocytes to assess this mechanism. In WT S49 cells, cAMP acts via protein kinase A (PKA) to induce G(1) phase cell cycle arrest and apoptosis. Treatment of WT and cAMP-Deathless (D-) S49 cells, which lack cAMP-promoted apoptosis, with the PKA agonist 8-(4-chlorophenylthio)-cAMP (CPT-cAMP) differentially regulates transcripts for numerous proapoptotic and antiapoptotic proteins. In contrast, kin-S49 cells (which lack PKA) show no cAMP-promoted changes in transcript expression. In this study, we use knockdown and overexpression approaches to define the role in cAMP/PKA-promoted apoptosis of the proapoptotic factor BIM (Bcl-2 interacting mediator of cell death), whose expression prominently increases in response to CPT-cAMP treatment of WT but not D- or kin- S49 cells. Conditional expression of BimL, one of the three major forms of Bim, increases apoptosis of WT, D-, and kin-S49 cells, whereas inhibition of cAMP-mediated induction of Bim isoforms by shRNAi attenuates CPT-cAMP-mediated apoptosis of WT S49 cells. Bim protein levels increase in subpopulations of CPT-cAMP-treated cells that undergo apoptosis. Thymic CD4(+)/CD8(+) cells isolated from Bim(-/-) mice corroborated the requirement of Bim expression for cAMP-promoted apoptosis. Thus, up-regulation of Bim appears to be an important determinant of cAMP/PKA-mediated apoptosis in immature T cells and may be a mechanism for such apoptosis in other cell types as well.
Collapse
Affiliation(s)
- Alexander C Zambon
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093, USA.
| | | | | | | |
Collapse
|