1
|
Siviero-Miachon AA, Lopes de Sousa AV, Simião BM, Araújo EO, Alvarenga R, Spinola-Castro AM, Longui CA. Glucocorticoid Sensitivity Among Young Survivors of Childhood Acute Lymphoblastic Leukemia: What Does It Matter? Horm Metab Res 2024; 56:718-726. [PMID: 39102841 DOI: 10.1055/a-2363-4823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
The aim of the study was to assess glucocorticoid sensitivity in survivors of childhood acute lymphoblastic leukemia using in vivo and in vitro tests. Thirty leukemia survivors of both sexes aged ≥18 years participated in the study and at least two years after therapy withdrawal. In vivo tests comprised: a) a very low dose intravenous dexamethasone suppression test for measurement of serum cortisol before, after, and % suppression, compared with 32 age-matched controls; and b) 0.25 mg overnight oral dexamethasone suppression test for assessment of salivary cortisol before, after, and % suppression. In vitro methods comprised: c) glucocorticoid receptor polymorphisms: BcI1-NR3C1 and A3669G; and d) splicing variant of glucocorticoid receptor GR-α mRNA by real-time quantitative polymerase chain reaction, compared with 32 controls. There was a reduction in salivary cortisol, and 73.3% of leukemia survivors showed high sensitivity according to % suppression after oral dexamethasone (p<0.05). Serum cortisol at baseline, after the test, % suppression after intravenous dexamethasone, and the percentage of high sensitivity were reduced in the leukemia group (%F=36.7; p<0.05). The BcI1-NR3C1 and A3669G polymorphisms were present in 11/30 (36.7%) and 5/30 (16.7%) patients, respectively. GR-α mRNA levels were lower in the leukemia group than in the controls (p<0.05). Survivors of acute lymphoblastic leukemia presented with reduced glucocorticoid sensitivity. Glucocorticoid sensitivity allows individualized treatment to avoid adverse effects and may be involved in cardiovascular disease risk among this particular group of cancer survivors.
Collapse
Affiliation(s)
- Adriana Aparecida Siviero-Miachon
- Pediatric Endocrinology, Federal University of Sao Paulo - UNIFESP/EPM, Sao Paulo, Brazil
- Pediatric Endocrinology, Hospital of the Support Group for Adolescent and Children with Cancer - GRAACC, Sao Paulo, Brazil
| | - Ana Virgínia Lopes de Sousa
- Pediatric Oncology, Hospital of the Support Group for Adolescent and Children with Cancer - GRAACC, Sao Paulo, Brazil
| | - Bruno Moreira Simião
- Pediatric Endocrinology, Federal University of Sao Paulo - UNIFESP/EPM, Sao Paulo, Brazil
| | - Elisangela Oliveira Araújo
- Physiological Sciences, Faculty of Medical Sciences of Santa Casa de Sao Paulo - FCM SCSP, Sao Paulo, Brazil
| | - Renato Alvarenga
- Physiological Sciences, Faculty of Medical Sciences of Santa Casa de Sao Paulo - FCM SCSP, Sao Paulo, Brazil
| | - Angela Maria Spinola-Castro
- Pediatric Endocrinology, Federal University of Sao Paulo - UNIFESP/EPM, Sao Paulo, Brazil
- Pediatric Endocrinology, Hospital of the Support Group for Adolescent and Children with Cancer - GRAACC, Sao Paulo, Brazil
| | - Carlos Alberto Longui
- Physiological Sciences, Faculty of Medical Sciences of Santa Casa de Sao Paulo - FCM SCSP, Sao Paulo, Brazil
| |
Collapse
|
2
|
Lockett J, Inder WJ, Clifton VL. The Glucocorticoid Receptor: Isoforms, Functions, and Contribution to Glucocorticoid Sensitivity. Endocr Rev 2024; 45:593-624. [PMID: 38551091 PMCID: PMC11244253 DOI: 10.1210/endrev/bnae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Indexed: 07/13/2024]
Abstract
Glucocorticoids exert pleiotropic effects on all tissues to regulate cellular and metabolic homeostasis. Synthetic forms are used therapeutically in a wide range of conditions for their anti-inflammatory benefits, at the cost of dose and duration-dependent side effects. Significant variability occurs between tissues, disease states, and individuals with regard to both the beneficial and deleterious effects. The glucocorticoid receptor (GR) is the site of action for these hormones and a vast body of work has been conducted understanding its function. Traditionally, it was thought that the anti-inflammatory benefits of glucocorticoids were mediated by transrepression of pro-inflammatory transcription factors, while the adverse metabolic effects resulted from direct transactivation. This canonical understanding of the GR function has been brought into question over the past 2 decades with advances in the resolution of scientific techniques, and the discovery of multiple isoforms of the receptor present in most tissues. Here we review the structure and function of the GR, the nature of the receptor isoforms, and the contribution of the receptor to glucocorticoid sensitivity, or resistance in health and disease.
Collapse
Affiliation(s)
- Jack Lockett
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Warrick J Inder
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Vicki L Clifton
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
| |
Collapse
|
3
|
Borin C, Pieters T, Serafin V, Ntziachristos P. Emerging Epigenetic and Posttranslational Mechanisms Controlling Resistance to Glucocorticoids in Acute Lymphoblastic Leukemia. Hemasphere 2023; 7:e916. [PMID: 37359189 PMCID: PMC10289758 DOI: 10.1097/hs9.0000000000000916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Glucocorticoids are extensively used for the treatment of acute lymphoblastic leukemia as they pressure cancer cells to undergo apoptosis. Nevertheless, glucocorticoid partners, modifications, and mechanisms of action are hitherto poorly characterized. This hampers our understanding of therapy resistance, frequently occurring in leukemia despite the current therapeutic combinations using glucocorticoids in acute lymphoblastic leukemia. In this review, we initially cover the traditional view of glucocorticoid resistance and ways of targeting this resistance. We discuss recent progress in our understanding of chromatin and posttranslational properties of the glucocorticoid receptor that might be proven beneficial in our efforts to understand and target therapy resistance. We discuss emerging roles of pathways and proteins such as the lymphocyte-specific kinase that antagonizes glucocorticoid receptor activation and nuclear translocation. In addition, we provide an overview of ongoing therapeutic approaches that sensitize cells to glucocorticoids including small molecule inhibitors and proteolysis-targeting chimeras.
Collapse
Affiliation(s)
- Cristina Borin
- Department of Biomolecular Medicine, Ghent University, Belgium
- Center for Medical Genetics, Ghent University and University Hospital, Belgium
- Cancer Research Institute Ghent (CRIG), Belgium
| | - Tim Pieters
- Department of Biomolecular Medicine, Ghent University, Belgium
- Center for Medical Genetics, Ghent University and University Hospital, Belgium
- Cancer Research Institute Ghent (CRIG), Belgium
| | - Valentina Serafin
- Department of Surgery Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Italy
| | - Panagiotis Ntziachristos
- Department of Biomolecular Medicine, Ghent University, Belgium
- Center for Medical Genetics, Ghent University and University Hospital, Belgium
- Cancer Research Institute Ghent (CRIG), Belgium
| |
Collapse
|
4
|
Wu S, Chen Y, Wang X, Weng S, Zhou W, Liu Z. Effect of EPA on Hsp90 and GRα protein expression in multiple myeloma drug-resistant cells. BMC Cancer 2021; 21:1076. [PMID: 34600510 PMCID: PMC8487534 DOI: 10.1186/s12885-021-08804-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Approximately 20% of MM patients harbor glucocorticoid (GC) resistance and are not responsive to therapeutic effect. Chaperoneheat-shock proteins Hsp90 is needed for ligand docking, The imbalance of Hsp90/GRα (glucocorticoid receptor α) may be an important cause of GC resistance. Recent studies have indicated that EPA could repress cancer cell growth by regulating critical influential factors in progression of cancer, consisting of resistance to drugs, chemosensitivity. The aim of the present study was to test the cytotoxic effects of EPA alone or EPA + Dexamethasone in dexamethasone-resistant MM cell (MM.1R) and investigate whether DHA can induce apoptosis and reverse acquired glucocorticoid resistance in dexamethasone-resistant MM cell (MM.1R). METHODS Cell Counting Kit-8 (CCK-8) was used to detect the proliferation of MM.1R cells after treating with EPA alone and EPA combined with DEX. Mitochondrial membrane potential was measured by flow cytometry and GRα and Hsp90 protein expression were assessed by western blot analysis. RESULTS EPA alone was able to inhibit cell proliferation as evidenced by CCK-8 assay and the tumor growth was remarkably suppressed by EPA + Dexamethasone, Cell apoptosis after EPA treatment was obviously observed by Flow cytometry analysis of the mitochondrial membrane potential. Analysis of Hsp90 and GRα proteins in MM.1R cells incubated with EPA revealed down-regulation of Hsp90 and up-regulation of GRα. Accordingly, the Hsp90/GRα ratio was significantly decreased with the increase of EPA concentration. CONCLUSIONS EPA might be used as a new effective treatment for reversal of glucocorticoid-resistance in multiple myeloma.
Collapse
Affiliation(s)
- Shenghao Wu
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China.
| | - Yuemiao Chen
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Xueshuang Wang
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Shanshan Weng
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Wenjin Zhou
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Zhen Liu
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| |
Collapse
|
5
|
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: 27] [Impact Index Per Article: 5.4] [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
|
6
|
Xie QM, Lou QY, Huang SW, Hu HQ, Li SS, Zhang M, Sun XX, Xu JH, Jiang SQ, Liu SX, Xu SQ, Cai J, Liu S, Pan FM, Tao JH, Qian L, Wang CH, Liang CM, Huang HL, Pan HF, Su H, Zou YF. Hsp70 Gene Polymorphisms Are Associated With Disease Susceptibility and HRQOL Improvement in Chinese Han Population With Systemic Lupus Erythematosus. J Clin Rheumatol 2020; 26:134-141. [PMID: 32453286 DOI: 10.1097/rhu.0000000000000986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of this study is to investigate whether heat shock protein 70 (Hsp70) gene polymorphisms are implicated in systemic lupus erythematous (SLE) susceptibility, the efficacy of glucocorticoids (GCs) treatment, and improvement of health-related quality of life. METHODS A total of 499 SLE patients and 499 controls were included in a case-control study, and 468 SLE patients treated with GCs for 12 weeks were involved in a follow-up study. Patients who completed the 12-week follow-up were divided into GCs-sensitive and GCs-insensitive group by using the SLE disease activity index. The SF-36 was used to evaluate the health-related quality of life of SLE patients, and genotyping was performed by improved multiplex ligation detection reaction. RESULTS rs2075800 was associated with SLE susceptibility (adjusted odds ratio [ORadj], 1.437; 95% confidence interval [CI], 1.113-1.855; Padj = 0.005; PBH = 0.020 by dominant model; ORadj, 1.602; 95% CI, 1.072-2.395; Padj = 0.022; PBH = 0.029 by TT vs CC model; ORadj = 1.396; 95% CI = 1.067-1.826; Padj = 0.015; PBH = 0.029 by TC vs CC model). In the follow-up study, rs2075799 was associated with the improvement in mental health (p = 0.004, PBH = 0.044), but we failed to find any association between the efficacy of GCs and Hsp70 gene polymorphisms. CONCLUSIONS Hsp70 gene polymorphisms may be associated with susceptibility to SLE and improvement of mental health in Chinese Han population.
Collapse
Affiliation(s)
- Qiao-Mei Xie
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Qiu-Yue Lou
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Shun-Wei Huang
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | | | - Su-Su Li
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Man Zhang
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Xiu-Xiu Sun
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Jian-Hua Xu
- Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University
| | | | - Sheng-Xiu Liu
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital of Anhui Medical University
| | - Sheng-Qian Xu
- Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University
| | - Jing Cai
- Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University
| | - Shuang Liu
- Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University
| | - Fa-Ming Pan
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Jin-Hui Tao
- Department of Rheumatology and Immunology, Anhui Medical University Affiliated Provincial Hospital
| | - Long Qian
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Anhui Medical University
| | - Chun-Huai Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Anhui Medical University
| | - Chun-Mei Liang
- Departments of Laboratory Medicine, School of Public Health
| | - Hai-Liang Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Hai-Feng Pan
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Hong Su
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| | - Yan-Feng Zou
- From the Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University
- The Key Laboratory of Anhui Medical Autoimmune Diseases
| |
Collapse
|
7
|
Ghoorun RA, Wu XH, Chen HL, Ren DL, Wu XB. Prognostic Significance of FKBP14 in Gastric Cancer. Onco Targets Ther 2019; 12:11567-11577. [PMID: 31920344 PMCID: PMC6941606 DOI: 10.2147/ott.s221943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/02/2019] [Indexed: 12/24/2022] Open
Abstract
Introduction Although our understanding on gastric cancer biology is better than a decade ago, its practical effect on screening and diagnosis remains limited. Moreover, there are no markers that can be accurately used in the clinic to diagnose early-stage gastric cancer or monitor the patient's response to therapy. Herein, we investigate whether FKBP14 is involved in the progression of gastric cancer. Methods The AGS cell line was chosen for over-expression analysis, whereas the SGC-7901 cell line was selected for knock-down analysis. AGS cells were transfected with an FKBP14 overexpression plasmid (AGS-PLV.O-FLAG). The expression pattern of FKBP14 in both cell lines was determined by Western blot and RT-PCR. Cell proliferation was assessed using Cell Counting Kit-8, whereas apoptosis was performed using flow cytometry. The expression of FKBP14 in 70 Chinese patients with gastric cancer was also investigated using tissue microarrays and compared with gastric cancer patients from The Cancer Genome Atlas. Results FKBP14 was highly expressed in SGC7901 and had a relatively low expression in AGS cells. Upregulation of FKBP14 in AGS cells promoted migration and invasion and inhibits apoptosis. Knock-down of FKBP14 resulted in a suppression in migration and invasion and promoted apoptosis in the SGC-7901 cell line. Effectively, gastric cancer patients had a higher expression of FKBP14, with a lower survival rate (P = 0.028). Patients with a high expression of FKBP14 were significantly correlated with lymph node metastasis (P =0.016), and an advanced histologic grade (P =0.021). Conclusion FKBP14 is often up-regulated in gastric cancer. Patients with a high expression of FKBP14 are usually associated with worse overall survival. FKBP14 is an oncogene in gastric cancer, and is a potential biomarker for GC diagnosis, invasion, and prognosis.
Collapse
Affiliation(s)
- Roshan Ara Ghoorun
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiao-Hua Wu
- Department of Oncology, Longhu People's Hospital, Shantou, Guangdong, People's Republic of China
| | - Hong-Lei Chen
- Department of Endoscopy, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, People's Republic of China
| | - Dong-Lin Ren
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiao-Bin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, People's Republic of China
| |
Collapse
|
8
|
Associations of HSP90AA2 gene polymorphisms with disease susceptibility, glucocorticoids efficacy and health-related quality of life in Chinese systemic lupus erythematosus patients. Genes Genomics 2018; 40:1069-1079. [PMID: 29907909 DOI: 10.1007/s13258-018-0714-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023]
Abstract
Although the current glucocorticoids (GCs) treatment for systemic lupus erythematosus (SLE) is effective to a certain extent, the difference in therapeutic effect between patients is still a widespread problem. Some patients can have repeated attacks that greatly diminish their quality of life. This study was conducted to investigate the relationship between HSP90AA2 polymorphisms and disease susceptibility, GCs efficacy and health-related quality of life (HRQoL) in Chinese SLE patients. A case-control study was performed in 470 SLE patients and 470 normal controls. Then, 444 patients in the case group were followed up for 12 weeks to observe efficacy of GCs and improvement of HRQoL. Two single nucleotide polymorphisms (SNPs) of HSP90AA2 were selected for genotyping: rs1826330 and rs6484340. HRQoL was assessed using the SF-36 questionnaire. The minor T allele of rs1826330 and the TT haplotype formed by rs1826330 and rs6484340 showed associations with decreased SLE risk (T allele: PBH = 0.022; TT haplotype: PBH = 0.033). A significant association between rs6484340 and improvement of HRQoL was revealed in the follow-up study. Five subscales of SF-36 were appeared to be influenced by rs6484340: total score of SF-36 (additive model: PBH = 0.026), physical function (additive model: PBH = 0.026), role-physical (recessive model: PBH = 0.041), mental health (dominant model: PBH = 0.047), and physical component summary (additive model: PBH = 0.026). No statistical significance was found between HSP90AA2 gene polymorphisms and GCs efficacy. These results revealed a genetic association between HSP90AA2 and SLE. Remarkably, HSP90AA2 has an impact on the improvement of HRQoL in Chinese population with SLE.
Collapse
|
9
|
Valenzuela CA, Zuloaga R, Mercado L, Einarsdottir IE, Björnsson BT, Valdés JA, Molina A. Chronic stress inhibits growth and induces proteolytic mechanisms through two different nonoverlapping pathways in the skeletal muscle of a teleost fish. Am J Physiol Regul Integr Comp Physiol 2017; 314:R102-R113. [PMID: 28978511 DOI: 10.1152/ajpregu.00009.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic stress detrimentally affects animal health and homeostasis, with somatic growth, and thus skeletal muscle, being particularly affected. A detailed understanding of the underlying endocrine and molecular mechanisms of how chronic stress affects skeletal muscle growth remains lacking. To address this issue, the present study assessed primary (plasma cortisol), secondary (key components of the GH/IGF system, muscular proteolytic pathways, and apoptosis), and tertiary (growth performance) stress responses in fine flounder ( Paralichthys adspersus) exposed to crowding chronic stress. Levels of plasma cortisol, glucocorticoid receptor 2 ( gr2), and its target genes ( klf15 and redd1) mRNA increased significantly only at 4 wk of crowding ( P < 0.05). The components of the GH/IGF system, including ligands, receptors, and their signaling pathways, were significantly downregulated at 7 wk of crowding ( P < 0.05). Interestingly, chronic stress upregulated the ubiquitin-proteasome pathway and the intrinsic apoptosis pathways at 4wk ( P < 0.01), whereas autophagy was only significantly activated at 7 wk ( P < 0.05), and meanwhile the ubiquitin-proteasome and the apoptosis pathways returned to control levels. Overall growth was inhibited in fish in the 7-wk chronic stress trial ( P < 0.05). In conclusion, chronic stress directly affects muscle growth and downregulates the GH/IGF system, an action through which muscular catabolic mechanisms are promoted by two different and nonoverlapping proteolytic pathways. These findings provide new information on molecular mechanisms involved in the negative effects that chronic stress has on muscle anabolic/catabolic signaling balance.
Collapse
Affiliation(s)
- Cristián A Valenzuela
- Universidad Andres Bello, Laboratorio de Biotecnología Molecular, Departamento de Ciencias Biológicas, Facultad Ciencias Biológicas , Santiago , Chile.,Interdisciplinary Center for Aquaculture Research , Concepción , Chile
| | - Rodrigo Zuloaga
- Universidad Andres Bello, Laboratorio de Biotecnología Molecular, Departamento de Ciencias Biológicas, Facultad Ciencias Biológicas , Santiago , Chile.,Interdisciplinary Center for Aquaculture Research , Concepción , Chile
| | - Luis Mercado
- Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Ingibjörg Eir Einarsdottir
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg , Gothenburg , Sweden
| | - Björn Thrandur Björnsson
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg , Gothenburg , Sweden
| | - Juan Antonio Valdés
- Universidad Andres Bello, Laboratorio de Biotecnología Molecular, Departamento de Ciencias Biológicas, Facultad Ciencias Biológicas , Santiago , Chile.,Interdisciplinary Center for Aquaculture Research , Concepción , Chile.,Universidad Andres Bello, Centro de Investigación Marina Quintay, Facultad de Ecología y Recursos Naturales , Valparaíso , Chile
| | - Alfredo Molina
- Universidad Andres Bello, Laboratorio de Biotecnología Molecular, Departamento de Ciencias Biológicas, Facultad Ciencias Biológicas , Santiago , Chile.,Interdisciplinary Center for Aquaculture Research , Concepción , Chile.,Universidad Andres Bello, Centro de Investigación Marina Quintay, Facultad de Ecología y Recursos Naturales , Valparaíso , Chile
| |
Collapse
|
10
|
Abstract
When individuals are exposed to stressful environmental challenges, the response varies widely in one or more of three components: psychology, behavior and physiology. This variability among individuals can be defined as temperament. In recent years, an increasing large body of evidence suggests that the dimensions of temperament, as well as personality, psychological disorders and behavioral traits, are influenced by genetic factors, and much of the variation appears to involve variation in genes or gene polymorphisms in the hypothalamic-pituitary-adrenocortical (HPA) axis and the behavior-controlling neurotransmitter networks. Here, we review our current understanding of the probabilistic impact of a number of candidate gene polymorphisms that control temperament, psychological disorders and behavioral traits in animals and human, including the gene polymorphisms related to corticotrophin-releasing hormone (CRH) production and adrenal cortisol production involved in the HPA axis, and a large number of gene polymorphisms in the dopaminergic and serotonergic neurotransmitter networks. It will very likely to assist in diagnosis and treatment of human relevant disorders, and provide useful contributions to our understanding of evolution, welfare and conservation, for animals in the wild and in production systems. Additionally, investigations of gene-gene and gene-environment complex interactions in humans and animals need further clear illustration.
Collapse
Affiliation(s)
- Xiaoyan Qiu
- a College of Animal Science and Technology, Southwest University , Chong Qing , PR China.,b UWA Institute of Agriculture and School of Animal Biology M082, Faculty of Sciences , University of Western Australia , Crawley , WA , Australia
| | - Graeme B Martin
- b UWA Institute of Agriculture and School of Animal Biology M082, Faculty of Sciences , University of Western Australia , Crawley , WA , Australia.,c Nuffield Department of Obstetrics and Gynecology , University of Oxford , Oxford , UK
| | - Dominique Blache
- b UWA Institute of Agriculture and School of Animal Biology M082, Faculty of Sciences , University of Western Australia , Crawley , WA , Australia
| |
Collapse
|
11
|
Song QQ, Xie WY, Tang YJ, Zhang J, Liu J. Genetic variation in the glucocorticoid pathway involved in interindividual differences in the glucocorticoid treatment. Pharmacogenomics 2017; 18:293-316. [PMID: 28112586 DOI: 10.2217/pgs-2016-0151] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) are widely used for treating asthma, rheumatoid arthritis, nephrotic syndrome, acute lymphoblastic leukemia and other autoimmune diseases. However, in a subgroup of patients, failure to respond to GCs is known as GC resistance or GC insensitivity. This represents an important barrier to effective treatment and a clinical problem requiring an urgent solution. Genetic variation in the GC pathway is a significant factor in interindividual differences in GC treatment. This article reviews the pharmacogenetics of GCs in diverse diseases based on the GC pathway.
Collapse
Affiliation(s)
- Qian-Qian Song
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Wan-Ying Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Yong-Jun Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| | - Jun Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Jie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, Hunan, P.R. China
| |
Collapse
|
12
|
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: 9.1] [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
|
13
|
Jackson RK, Irving JAE, Veal GJ. Personalization of dexamethasone therapy in childhood acute lymphoblastic leukaemia. Br J Haematol 2016; 173:13-24. [DOI: 10.1111/bjh.13924] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Rosanna K. Jackson
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| | - Julie A. E. Irving
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| | - Gareth J. Veal
- Northern Institute for Cancer Research; Newcastle University; Newcastle upon Tyne UK
| |
Collapse
|
14
|
Milani M, Laranjeira ABA, de Vasconcellos JF, Brandalise SR, Nowill AE, Yunes JA. Plasma Hsp90 Level as a Marker of Early Acute Lymphoblastic Leukemia Engraftment and Progression in Mice. PLoS One 2015; 10:e0129298. [PMID: 26068922 PMCID: PMC4466233 DOI: 10.1371/journal.pone.0129298] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/08/2015] [Indexed: 11/29/2022] Open
Abstract
Current monitoring of acute lymphoblastic leukemia (ALL) in living mice is based on FACS analysis of blood hCD45+ cells. In this work, we evaluated the use of human IGFBP2, B2M or Hsp90 as soluble markers of leukemia. ELISA for B2M and IGFBP2 resulted in high background levels in healthy animals, precluding its use. Conversely, plasma levels of Hsp90 showed low background and linear correlation to FACS results. In another experiment, we compared Hsp90 levels with percentage of hCD45+ cells in blood, bone marrow, liver and spleen of animals weekly sacrificed. Hsp90 levels proved to be a superior method for the earlier detection of ALL engraftment and correlated linearly to ALL burden and progression in all compartments, even at minimal residual disease levels. Importantly, the Hsp90/hCD45+ ratio was not altered when animals were treated with dexamethasone or a PI3K inhibitor, indicating that chemotherapy does not directly interfere with leukemia production of Hsp90. In conclusion, plasma Hsp90 was validated as a soluble biomarker of ALL, useful for earlier detection of leukemia engraftment, monitoring leukemia kinetics at residual disease levels, and pre-clinical or mouse avatar evaluations of anti-leukemic drugs.
Collapse
Affiliation(s)
- Mateus Milani
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP, Brazil
| | | | | | | | - Alexandre Eduardo Nowill
- Centro Integrado de Pesquisas Oncohematologicas da Infância (CIPOI), Faculdade de Ciências Médicas (FCM), Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - José Andrés Yunes
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP, Brazil
- Departamento de Genética Médica, FCM, UNICAMP, Campinas, SP, Brazil
- * E-mail:
| |
Collapse
|
15
|
Sun Z, Dong J, Zhang S, Hu Z, Cheng K, Li K, Xu B, Ye M, Nie Y, Fan D, Zou H. Identification of chemoresistance-related cell-surface glycoproteins in leukemia cells and functional validation of candidate glycoproteins. J Proteome Res 2014; 13:1593-601. [PMID: 24467213 DOI: 10.1021/pr4010822] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemoresistance remains the most significant obstacle to successful chemotherapy for leukemia, and its exact mechanism is still unknown. In this work, we used the cell-surface capturing method together with quantitative proteomics to investigate differences in the glycoproteomes of adriamycin-sensitive and adriamycin-resistant leukemia cells. Two quantitative methods, isotopic dimethyl labeling and SWATH, were used to quantify glycoproteins, and 35 glycoproteins were quantified by both methods. High correlation was observed between the glycoproteins quantified by the above two methods, and 15 glycoproteins displayed a consistent significant change trend in both sets of quantitative results. These 15 proteins included classical multidrug resistance-related glycoproteins such as ABCB1 as well as a set of novel glycoproteins that have not previously been reported to be associated with chemoresistance in leukemia cells. Further validation with quantitative real-time PCR and Western blotting confirmed the proteomic screening results. Subsequent functional experiments based on RNA interference technology showed that CTSD, FKBP10, and SLC2A1 are novel genes that participate in the acquisition and maintenance of the adriamycin-resistant phenotype in leukemia cells.
Collapse
Affiliation(s)
- Zhen Sun
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences , 457 Zhongshan Road, Dalian 116023, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Dominance of the strongest: inflammatory cytokines versus glucocorticoids. Cytokine Growth Factor Rev 2013; 25:21-33. [PMID: 24412262 DOI: 10.1016/j.cytogfr.2013.12.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/15/2013] [Indexed: 02/08/2023]
Abstract
Pro-inflammatory cytokines are involved in the pathogenesis of many inflammatory diseases, and the excessive expression of many of them is normally counteracted by glucocorticoids (GCs), which are steroids that bind to the glucocorticoid receptor (GR). Hence, GCs are potent inhibitors of inflammation, and they are widely used to treat inflammatory diseases, such as asthma, rheumatoid arthritis and inflammatory bowel disease. However, despite the success of GC therapy, many patients show some degree of GC unresponsiveness, called GC resistance (GCR). This is a serious problem because it limits the full therapeutic exploitation of the anti-inflammatory power of GCs. Patients with reduced GC responses often have higher cytokine levels, and there is a complex interplay between GCs and cytokines: GCs downregulate pro-inflammatory cytokines while cytokines limit GC action. Treatment of inflammatory diseases with GCs is successful when GCs dominate. But when cytokines overrule the anti-inflammatory actions of GCs, patients become GC insensitive. New insights into the molecular mechanisms of GR-mediated actions and GCR are needed for the design of more effective GC-based therapies.
Collapse
|
17
|
Yang N, Ray DW, Matthews LC. Current concepts in glucocorticoid resistance. Steroids 2012; 77:1041-9. [PMID: 22728894 DOI: 10.1016/j.steroids.2012.05.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/24/2012] [Accepted: 05/29/2012] [Indexed: 01/04/2023]
Abstract
Glucocorticoids (GCs) are the most potent anti-inflammatory agents known. A major factor limiting their clinical use is the wide variation in responsiveness to therapy. The high doses of GC required for less responsive patients means a high risk of developing very serious side effects. Variation in sensitivity between individuals can be due to a number of factors. Congenital, generalized GC resistance is very rare, and is due to mutations in the glucocorticoid receptor (GR) gene, the receptor that mediates the cellular effects of GC. A more common problem is acquired GC resistance. This localized, disease-associated GC resistance is a serious therapeutic concern and limits therapeutic response in patients with chronic inflammatory disease. It is now believed that localized resistance can be attributed to changes in the cellular microenvironment, as a consequence of chronic inflammation. Multiple factors have been identified, including alterations in both GR-dependent and -independent signaling downstream of cytokine action, oxidative stress, hypoxia and serum derived factors. The underlying mechanisms are now being elucidated, and are discussed here. Attempts to augment tissue GC sensitivity are predicted to permit safe and effective use of low-dose GC therapy in inflammatory disease.
Collapse
Affiliation(s)
- Nan Yang
- Endocrine Sciences Research Group, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
18
|
Bhadri VA, Trahair TN, Lock RB. Glucocorticoid resistance in paediatric acute lymphoblastic leukaemia. J Paediatr Child Health 2012; 48:634-40. [PMID: 22050419 DOI: 10.1111/j.1440-1754.2011.02212.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Glucocorticoids (GCs), such as prednisolone and dexamethasone, are key components in multi-agent chemotherapy protocols used for the treatment of acute lymphoblastic leukaemia (ALL). Approximately 10% of children with ALL will respond poorly to GCs, and GC resistance is associated with a significantly inferior outcome. This review summarises the current knowledge of GC resistance in ALL, including the roles of the GC receptor and its co-chaperone molecules, the pro-apoptotic and pro-survival B-cell lymphoma 2 family members and alternative non-apoptotic mechanisms of cell death. It concludes with a discussion on therapeutic attempts to overcome GC resistance.
Collapse
Affiliation(s)
- Vivek A Bhadri
- Centre for Children's Cancer and Blood Disorders, Sydney Children's Hospital, Randwick, NSW 2031, Australia.
| | | | | |
Collapse
|
19
|
Haskins WE, Eedala S, Jadhav YA, Labhan MS, Pericherla VC, Perlman EJ. Insights on neoplastic stem cells from gel-based proteomics of childhood germ cell tumors. Pediatr Blood Cancer 2012; 58:722-8. [PMID: 21793190 PMCID: PMC3204330 DOI: 10.1002/pbc.23282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 06/22/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Childhood germ cell tumors (cGCTs), believed to arise from transformed primordial germ cells by an unknown mechanism, provide a unique model system for investigating cell signaling, pluripotency, and the microenvironment of neoplastic stem cells (NSCs) in vivo. This is the first report of proteomics of cGCTs. PROCEDURE Four dysgerminomas (DYSs) and four childhood endodermal sinus tumors (cESTs), resembling self-renewing and differentiating NSCs, respectively, were selected. Proteomic studies were performed by 2-DE, SDS-PAGE, and cLC/MS/MS with protein database searching. RESULTS 2-DE: 9 of 941 spots were differentially regulated with greater than a twofold change in spot volume for at least three of four gels in each group. Two of nine spots had P values for the t-test analysis of comparisons less than 0.001, while the remaining spots had P values from 0.013 to 0.191. Top-ranked proteins were identified in nine of nine spots with 4.0-38% sequence coverage. APOA1, CRK, and PDIA3 were up-regulated in cESTs. TFG, TYMP, VCP, RBBP, FKBP4, and BiP were up-regulated in DYSs. SDS-PAGE: Up-regulation of NF45 and FKBP4 was observed in four of four cESTs and DYSs, respectively. The fold-changes observed correspond with characteristic genetic changes. CONCLUSION Differential regulation of FKBP4 and NF45, combined with previous research on immunosuppressant binding, suggests that glucocorticoid receptor signaling merits further investigation in cGCTs and NSCs.
Collapse
Affiliation(s)
- William E. Haskins
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Medicine, Division of Hematology & Medical Oncology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229,Correspondence: William E. Haskins, Ph.D., Dept. of Biology-BSE 3.108A, The University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249-0662, , Phone: (210)563-4492, Fax: (210)458-5658
| | - Sruthi Eedala
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Y.L. Avinash Jadhav
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Manbir S. Labhan
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Vidya C. Pericherla
- Pediatric Biochemistry Laboratory, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, Department of RCMI Proteomics, University of Texas at San Antonio, San Antonio, TX, 78249, Department of Protein Biomarkers Cores, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Interdisciplinary Health Research, University of Texas at San Antonio, San Antonio, TX, 78249, Center for Research & Training in the Sciences, University of Texas at San Antonio, San Antonio, TX, 78249
| | - Elizabeth J. Perlman
- Department of Pathology, Northwestern University’s Feinberg School of Medicine and Robert H. Lurie Cancer Center, Chicago, IL, 60614
| |
Collapse
|
20
|
Gross KL, Lu NZ, Cidlowski JA. Molecular mechanisms regulating glucocorticoid sensitivity and resistance. Mol Cell Endocrinol 2009; 300:7-16. [PMID: 19000736 PMCID: PMC2674248 DOI: 10.1016/j.mce.2008.10.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 10/08/2008] [Accepted: 10/08/2008] [Indexed: 02/07/2023]
Abstract
Glucocorticoid receptor agonists are mainstays in the treatment of various malignancies of hematological origin. Glucocorticoids are included in therapeutic regimens for their ability to stimulate intracellular signal transduction cascades that culminate in alterations in the rate of transcription of genes involved in cell cycle progression and programmed cell death. Unfortunately, subpopulations of patients undergoing systemic glucocorticoid therapy for these diseases are or become insensitive to glucocorticoid-induced cell death, a phenomenon recognized as glucocorticoid resistance. Multiple factors contributing to glucocorticoid resistance have been identified. Here we summarize several of these mechanisms and describe the processes involved in generating a host of glucocorticoid receptor isoforms from one gene. The potential role of glucocorticoid receptor isoforms in determining cellular responsiveness to glucocorticoids is emphasized.
Collapse
Affiliation(s)
- Katherine L. Gross
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Drive, Research Triangle Park, North Carolina 27709, USA
| | - Nick Z. Lu
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Drive, Research Triangle Park, North Carolina 27709, USA
| | - John A. Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Drive, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
21
|
Becker H, Sturm A, Bron JE, Schirmer K, Bury NR. The A/B domain of the teleost glucocorticoid receptors influences partial nuclear localization in the absence of hormone. Endocrinology 2008; 149:4567-76. [PMID: 18483153 DOI: 10.1210/en.2007-1683] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The glucocorticoid (GR) and mineralocorticoid receptor (MR) of extant jawed vertebrates emerged after duplication of an ancestral corticosteroid receptor. The ancestral corticosteroid receptor resembled extant MRs in hormone selectivity, and the different ligand specificity of extant GRs is a secondary derived characteristic. An additional characteristic that distinguishes the mammalian GR from the MR is the cellular distribution pattern in the absence of hormone: the naïve GR resides in the cytoplasm, whereas the naïve MR is found in both the nucleus and cytoplasm. Our results show, by the use of green fluorescent protein-tagged fusion proteins, that the GRs [rainbow trout (rt) GR1 and rtGR2] from a lower vertebrate, the teleost fish, rainbow trout (Oncorhynchus mykiss) resemble mammalian MR rather than GR in their subcellular localization pattern. The addition of cortisol caused the remaining cytoplasmic rtGR1 and rtGR2 to migrate to the nucleus. The speed of nuclear localization was cortisol concentration dependent, with rtGR2 being more sensitive than rtGR1, mimicking the transactivational properties of the receptors in which the cortisol EC50 value is an order of magnitude lower for rtGR2. By the use of chimera constructs between the trout GRs and the rat GR C656G, we show that the E domain of the trout receptors are not involved in the nucleocytoplasmic localization of naïve trout GRs, but the A/B domain, especially if linked to the corresponding trout CD region, plays a pivotal role in the cellular distribution pattern. This is unrelated to the difference in the trout GRs transactivation sensitivity, which is determined by the receptor's E-domains.
Collapse
Affiliation(s)
- Heidi Becker
- Nutritional Sciences Research Division, King's College London, London SE1 9NH, United Kingdom
| | | | | | | | | |
Collapse
|
22
|
Wang SS, Kamphuis W, Huitinga I, Zhou JN, Swaab DF. Gene expression analysis in the human hypothalamus in depression by laser microdissection and real-time PCR: the presence of multiple receptor imbalances. Mol Psychiatry 2008; 13:786-99, 741. [PMID: 18427561 DOI: 10.1038/mp.2008.38] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hyperactivity of corticotropin-releasing factor (CRF) neurons in the paraventricular nucleus (PVN) of the hypothalamus is a prominent feature in depression and may be important in the etiology of this disease. The activity of the CRF neurons in the stress response is modulated by a number of factors that stimulate or inhibit CRF expression, including (1) corticosteroid receptors and their chaperones, heat shock proteins 70 and 90, (2) sex hormone receptors, (3) CRF receptors 1 (CRFR1) and 2, (4) cytokines interleukin 1-beta and tumor necrosis factor-alpha, (5) neuropeptides and receptors, vasopressin (AVP), AVP receptor 1a (AVPR1A) and oxytocin and (6) transcription factor cAMP-response element-binding protein. We hypothesized that, in depression, the transcript levels of those genes that are involved in the activation of the hypothalamo-pituitary-adrenal (HPA) axis are upregulated, whereas the transcript levels of the genes involved in the inhibition of the HPA axis are downregulated. We performed laser microdissection and real-time PCR in the PVN and as a control in the supraoptic nucleus. Snap-frozen post-mortem hypothalami of seven depressed and seven matched controls were used. We found significantly increased CRF mRNA levels in the PVN of the depressed patients. This was accompanied by a significantly increased expression of four genes that are involved in the activation of CRF neurons, that is, CRFR1, estrogen receptor-alpha, AVPR1A and mineralocorticoid receptor, while the expression of the androgen receptor mRNA involved in the inhibition of CRF neurons was decreased significantly. These findings raise the possibility that a disturbed balance in the production of receptors may contribute to the activation of the HPA axis in depression.
Collapse
Affiliation(s)
- S-S Wang
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
23
|
Bury NR, Sturm A. Evolution of the corticosteroid receptor signalling pathway in fish. Gen Comp Endocrinol 2007; 153:47-56. [PMID: 17470371 DOI: 10.1016/j.ygcen.2007.03.009] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 03/11/2007] [Accepted: 03/19/2007] [Indexed: 11/21/2022]
Abstract
The corticosteroid receptors (CR) control a vast array of physiological processes acting primarily as ligand-dependent transcription factors. The origins of the gnathostomata CRs can be traced back to an ancestral steroid receptor present in a primitive agnathan vertebrate. A genome duplication event in the early gnathostomes is believed to have produced a set of two CRs still present today in Sarcopterygii (lobe-finned fish and tetrapods), i.e. a glucocorticoid (GR) and mineralocorticoid receptor (MR), with divergent function and different ligands, cortisol and aldosterone, respectively. A further genome duplication occurred in the early evolutionary history of the teleosts, and the teleost CR system seems to have diversified, consisting now of 2 GRs and a MR. Teleosts lack aldosterone and the main corticosteroid is believed to be cortisol. However, the mineralocorticoid, 11-deoxycorticosterone (DOC), has been identified as an agonist for the rainbow trout MR, suggesting it may be the ancestral ligand for the MR. The retention of two GRs in teleosts suggests neofunctionalisation of one of the duplicated genes, but this hypothesis requires further work. In rainbow trout, transactivation and transrepression activities of the two GRs show marked differences in their sensitivity to glucocorticoids, suggesting a mechanism that may allow the two GRs to control different physiological pathways. Whether a similar mechanism is seen throughout the actinopterygii or whether this is specific to the salmonid lineage remains to be verified.
Collapse
Affiliation(s)
- Nicolas R Bury
- Department of Biochemistry, Nutritional Sciences Research Group, Franklin Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | | |
Collapse
|
24
|
Bornhauser BC, Bonapace L, Lindholm D, Martinez R, Cario G, Schrappe M, Niggli FK, Schäfer BW, Bourquin JP. Low-dose arsenic trioxide sensitizes glucocorticoid-resistant acute lymphoblastic leukemia cells to dexamethasone via an Akt-dependent pathway. Blood 2007; 110:2084-91. [PMID: 17537996 DOI: 10.1182/blood-2006-12-060970] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Incorporation of apoptosis-inducing agents into current therapeutic regimens is an attractive strategy to improve treatment for drug-resistant leukemia. We tested the potential of arsenic trioxide (ATO) to restore the response to dexamethasone in glucocorticoid (GC)-resistant acute lymphoblastic leukemia (ALL). Low-dose ATO markedly increased in vitro GC sensitivity of ALL cells from T-cell and precursor B-cell ALL patients with poor in vivo response to prednisone. In GC-resistant cell lines, this effect was mediated, at least in part, by inhibition of Akt and affecting downstream Akt targets such as Bad, a proapoptotic Bcl-2 family member, and the X-linked inhibitor of apoptosis protein (XIAP). Combination of ATO and dexamethasone resulted in increased Bad and rapid down-regulation of XIAP, while levels of the antiapoptotic regulator Mcl-1 remained unchanged. Expression of dominant-active Akt, reduction of Bad expression by RNA interference, or overexpression of XIAP abrogated the sensitizing effect of ATO. The inhibitory effect of XIAP overexpression was reduced when the Akt phosphorylation site was mutated (XIAP-S87A). These data suggest that the combination of ATO and glucocorticoids could be advantageous in GC-resistant ALL and reveal additional targets for the evaluation of new antileukemic agents.
Collapse
Affiliation(s)
- Beat C Bornhauser
- Department of Oncology, Children's Hospital, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bachmann PS, Gorman R, Papa RA, Bardell JE, Ford J, Kees UR, Marshall GM, Lock RB. Divergent Mechanisms of Glucocorticoid Resistance in Experimental Models of Pediatric Acute Lymphoblastic Leukemia. Cancer Res 2007; 67:4482-90. [PMID: 17483364 DOI: 10.1158/0008-5472.can-06-4244] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cell line models of glucocorticoid resistance in childhood acute lymphoblastic leukemia (ALL) almost invariably exhibit altered glucocorticoid receptor (GR) function. However, these findings are incongruous with those using specimens derived directly from leukemia patients, in which GR alterations are rarely found. Consequently, mechanisms of glucocorticoid resistance in the clinical setting remain largely unresolved. We present a novel paradigm of glucocorticoid resistance in childhood ALL, in which patient biopsies have been directly established as continuous xenografts in immune-deficient mice, without prior in vitro culture. We show that the GRs from six highly dexamethasone-resistant xenografts (in vitro IC(50) >10 micromol/L) exhibit no defects in ligand-induced nuclear translocation and binding to a consensus glucocorticoid response element (GRE). This finding contrasts with five commonly used leukemia cell lines, all of which exhibited defective GRE binding. Moreover, whereas the GRs of dexamethasone-resistant xenografts were transcriptionally active, as assessed by the ability to induce the glucocorticoid-induced leucine zipper (GILZ) gene, resistance was associated with failure to induce the bim gene, which encodes a proapoptotic BH3-only protein. Furthermore, the receptor tyrosine kinase inhibitor, SU11657, completely reversed dexamethasone resistance in a xenograft expressing functional GR, indicating that pharmacologic reversal of glucocorticoid resistance in childhood ALL is achievable.
Collapse
Affiliation(s)
- Petra S Bachmann
- Children's Cancer Institute Australia for Medical Research, The University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Shi ZZ, Zhang JW, Zheng S. What we know about ST13, a co-factor of heat shock protein, or a tumor suppressor? J Zhejiang Univ Sci B 2007; 8:170-6. [PMID: 17323428 PMCID: PMC1810382 DOI: 10.1631/jzus.2007.b0170] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This article is to summarize the molecular and functional analysis of the gene "suppression of tumorigenicity 13" (ST13). ST13 is in fact the gene encoding Hsp70 interacting protein (Hip), a co-factor (co-chaperone) of the 70-kDa heat shock proteins (Hsc/Hsp70). By collaborating with other positive co-factors such as Hsp40 and the Hsp70-Hsp90 organizing protein (Hop), or competing with negative co-factors such as Bcl2-associated athanogen 1 (Bag1), Hip facilitates may facilitate the chaperone function of Hsc/Hsp70 in protein folding and repair, and in controlling the activity of regulatory proteins such as steroid receptors and regulators of proliferation or apoptosis. Although the nomenclature of ST13 implies a role in the suppression of tumorigenicity (ST), to date available experimental data are not sufficient to support its role in cancer development, except for the possible down-regulation of ST13 in gastric and colorectal cancers. Further investigation of this gene at the physiological level would benefit our understanding of diseases such as endocrinological disorders, cancer, and neurodegeneration commonly associated with protein misfolding.
Collapse
|
27
|
Lauten M, Schrauder A, Kardinal C, Harbott J, Welte K, Schlegelberger B, Schrappe M, von Neuhoff N. Unsupervised proteome analysis of human leukaemia cells identifies the Valosin-containing protein as a putative marker for glucocorticoid resistance. Leukemia 2006; 20:820-6. [PMID: 16541142 DOI: 10.1038/sj.leu.2404162] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The response to initial glucocorticoid therapy in childhood acute lymphoblastic leukaemia (ALL) reliably predicts the response to multiagent chemotherapy. Patients resistant to glucocorticoids (prednisone poor responders (PPR)) have a poorer event-free survival compared to glucocorticoid-sensitive patients (prednisone good responders (PGR)). A case-control study was performed to investigate differential protein expression in leukaemic blasts from PGR and PPR childhood ALL patients. Two-dimensional gel electrophoresis (2-DE) was used for an unsupervised screening and surface enhanced laser desorption/ionisation-time of flight mass spectrometry (SELDI-TOF MS) for the characterisation of protein spots. In difference maps of average gels for the proteomes of each responder group, differentially expressed proteins were identified after tryptic digestion and spotting onto H4-SELDI-TOF-MS chips. Proteins overexpressed in PPR were Catalase, RING finger protein 22 alpha, Valosin-containing protein (VCP) and a G-protein-coupled receptor. Proteins overexpressed in PGR were protein kinase C and malate dehydrogenase. Valosin-containing protein was chosen for validation and quantification by Western blot analysis in a second case-control group of ALL patients. In this second independent cohort, median VCP expression (P25-P75) was 0.15 (0.11-0.28) in PGR and 0.34 (0.14-0.99) in PPR patients (P = 0.04). We conclude that high VCP expression is associated with poor prednisone response in childhood ALL patients.
Collapse
Affiliation(s)
- M Lauten
- Department of Paediatrics and Adolescent Medicine, Division of Paediatric Haematology and Oncology, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ziino O, D'Urbano LE, De Benedetti F, Conter V, Barisone E, De Rossi G, Basso G, Aricò M. The MIF-173G/C polymorphism does not contribute to prednisone poor response in vivo in childhood acute lymphoblastic leukemia. Leukemia 2006; 19:2346-7. [PMID: 16208413 DOI: 10.1038/sj.leu.2403973] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
29
|
Current Awareness in Hematological Oncology. Hematol Oncol 2005. [DOI: 10.1002/hon.730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Murphy PJM. Regulation of glucocorticoid receptor steroid binding and trafficking by the hsp90/hsp70-based chaperone machinery: implications for clinical intervention. Leukemia 2005; 19:710-2. [PMID: 15759036 DOI: 10.1038/sj.leu.2403687] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|