51
|
Wang Z, Belghasem M, Salih E, Henderson J, Igwebuike C, Havasi A, Borkan SC. T95 nucleophosmin phosphorylation as a novel mediator and marker of regulated cell death in acute kidney injury. Am J Physiol Renal Physiol 2020; 319:F552-F561. [PMID: 32686519 PMCID: PMC7509286 DOI: 10.1152/ajprenal.00230.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/29/2020] [Accepted: 07/09/2020] [Indexed: 12/25/2022] Open
Abstract
The function of site-specific phosphorylation of nucleophosmin (NPM), an essential Bax chaperone, in stress-induced cell death is unknown. We hypothesized that NPM threonine 95 (T95) phosphorylation both signals and promotes cell death. In resting cells, NPM exclusively resides in the nucleus and T95 is nonphosphorylated. In contrast, phosphorylated T95 NPM (pNPM T95) accumulates in the cytosol after metabolic stress, in multiple human cancer cell lines following γ-radiation, and in postischemic human kidney tissue. Based on the T95 phosphorylation consensus sequence, we hypothesized that glycogen synthase kinase-3β (GSK-3β) regulates cytosolic NPM translocation by phosphorylating T95 NPM. In a cell-free system, GSK-3β phosphorylated a synthetic NPM peptide containing T95. In vitro, bidirectional manipulation of GSK-3β activity substantially altered T95 phosphorylation, cytosolic NPM translocation, and cell survival during stress, mechanistically linking these lethal events. Furthermore, GSK-3β inhibition in vivo decreased cytosolic pNPM T95 accumulation in kidney tissue after experimental ischemia. In patients with acute kidney injury, both cytosolic NPM accumulation in proximal tubule cells and NPM-rich intratubular casts were detected in frozen renal biopsy tissue. These observations show, for the first time, that GSK-3β promotes cell death partly by phosphorylating NPM at T95, to promote cytosolic NPM accumulation. T95 NPM is also a rational therapeutic target to ameliorate ischemic renal cell injury and may be a universal injury marker in mammalian cells.
Collapse
Affiliation(s)
- Zhiyong Wang
- Renal Section, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Mostafa Belghasem
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts
| | - Erdjan Salih
- Goldman School of Dentistry, Boston University, Boston, Massachusetts
| | - Joel Henderson
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts
| | - Chinaemere Igwebuike
- Renal Section, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Andrea Havasi
- Renal Section, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| | - Steven C Borkan
- Renal Section, Department of Medicine, Boston Medical Center, Boston University, Boston, Massachusetts
| |
Collapse
|
52
|
Cela I, Di Matteo A, Federici L. Nucleophosmin in Its Interaction with Ligands. Int J Mol Sci 2020; 21:E4885. [PMID: 32664415 PMCID: PMC7402337 DOI: 10.3390/ijms21144885] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022] Open
Abstract
Nucleophosmin (NPM1) is a mainly nucleolar protein that shuttles between nucleoli, nucleoplasm and cytoplasm to fulfill its many functions. It is a chaperone of both nucleic acids and proteins and plays a role in cell cycle control, centrosome duplication, ribosome maturation and export, as well as the cellular response to a variety of stress stimuli. NPM1 is a hub protein in nucleoli where it contributes to nucleolar organization through heterotypic and homotypic interactions. Furthermore, several alterations, including overexpression, chromosomal translocations and mutations are present in solid and hematological cancers. Recently, novel germline mutations that cause dyskeratosis congenita have also been described. This review focuses on NPM1 interactions and inhibition. Indeed, the list of NPM1 binding partners is ever-growing and, in recent years, many studies contributed to clarifying the structural basis for NPM1 recognition of both nucleic acids and several proteins. Intriguingly, a number of natural and synthetic ligands that interfere with NPM1 interactions have also been reported. The possible role of NPM1 inhibitors in the treatment of multiple cancers and other pathologies is emerging as a new therapeutic strategy.
Collapse
Affiliation(s)
- Ilaria Cela
- Center for Advanced Studies and Technology (CAST), University of Chieti “G. d’Annunzio”, Via Polacchi, 66100 Chieti, Italy;
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti “G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| | - Adele Di Matteo
- Institute of Molecular Biology and Pathology (IBPM) of the CNR, c/o “Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Roma, Italy;
| | - Luca Federici
- Center for Advanced Studies and Technology (CAST), University of Chieti “G. d’Annunzio”, Via Polacchi, 66100 Chieti, Italy;
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti “G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
53
|
Chen B, Li Y, Nie Y, Tang A, Zhou Q. Long non-coding RNA LINC01268 promotes cell growth and inhibits cell apoptosis by modulating miR-217/SOS1 axis in acute myeloid leukemia. ACTA ACUST UNITED AC 2020; 53:e9299. [PMID: 32609259 PMCID: PMC7326380 DOI: 10.1590/1414-431x20209299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
Abstract
The aim of this study was to evaluate the pathogenic role of newly identified long non-coding (lnc)-RNA LINCO1268 in acute myeloid leukemia (AML), and investigate its therapeutic potential. The expression level of LINC01268 in AML was measured by quantitative PCR (qPCR). The viability, cell cycle progression, and apoptosis of AML cells were measured by CCK-8 assay and flow cytometry, respectively. The interaction between LINC01268 and miR-217 were predicted by the miRDB website, and then verified by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. The relationship between miR-217 and SOS1 was predicted by TargetScan website, and verified by luciferase reporter assay. LINC01268 was significantly upregulated by 1.6 fold in bone marrow samples of AML patients, which was associated with poor prognosis. LINC01268 was also significantly upregulated in AML cells. LINC01268 knockdown inhibited viability and cell cycle progression but promoted apoptosis of AML cells. Furthermore, LINC01268 functioned as a ceRNA via competitively binding to miR-217, and SOS1 was identified as a target of miR-217. Moreover, LINC01268 positively regulated SOS1 expression to promote AML cell viability and cell cycle progression but inhibited apoptosis via sponging miR-217. LINC01268 promoted cell growth and inhibited cell apoptosis through modulating miR-217/SOS1 axis in AML. This study offers a novel molecular mechanism for a better understanding of the pathology of AML. LINC01268 could be considered as a potential biomarker for the therapy and diagnosis of AML.
Collapse
Affiliation(s)
- Beili Chen
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Yuchuan Li
- Department of Gynecology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Yuwei Nie
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Ailin Tang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Qin Zhou
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
54
|
Mannan A, Muhsen IN, Barragán E, Sanz MA, Mohty M, Hashmi SK, Aljurf M. Genotypic and Phenotypic Characteristics of Acute Promyelocytic Leukemia Translocation Variants. Hematol Oncol Stem Cell Ther 2020; 13:189-201. [PMID: 32473106 DOI: 10.1016/j.hemonc.2020.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
Acute promyelocytic leukemia (APL) is a special disease entity of acute myeloid leukemia (AML). The clinical use of all-trans retinoic acid (ATRA) has transformed APL into the most curable form of AML. The majority of APL cases are characterized by the fusion gene PML-RARA. Although the PML-RARA fusion gene can be detected in almost all APL cases, translocation variants of APL have been reported. To date, this is the most comprehensive review of these translocations, discussing 15 different variants. Reviewed genes involved in APL variants include: ZBTB16, NPM, NuMA, STAT5b, PRKAR1A, FIP1L1, BCOR, NABP1, TBLR1, GTF2I, IRF2BP2, FNDC3B, ADAMDTS17, STAT3, and TFG. The genotypic and phenotypic features of APL translocations are summarized. All reported studies were either case reports or case series indicating the rarity of these entities and limiting the ability to drive conclusions regarding their characteristics. However, reported variants have shown variable clinical and morphological features, with diverse responsiveness to ATRA.
Collapse
Affiliation(s)
- Abdul Mannan
- Betsi Cadwaladr University Health Board, Bangor, UK
| | - Ibrahim N Muhsen
- Department of Medicine, Houston Methodist Hospital, Houston, TX, USA.
| | - Eva Barragán
- Department of Hematology, Hospital Universitari i Politecnic La Fe, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
| | - Miguel A Sanz
- Department of Hematology, Hospital Universitari i Politecnic La Fe, Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
| | | | - Shahrukh K Hashmi
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
55
|
Aref S, Azmy E, Ibrahim L, Sabry M, Agdar ME. Prognostic value of CD25/CD123 pattern of expression in acute myeloid leukemia patients with normal cytogenetic. Leuk Res Rep 2020; 13:100203. [PMID: 32514390 PMCID: PMC7267724 DOI: 10.1016/j.lrr.2020.100203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/23/2020] [Accepted: 05/10/2020] [Indexed: 01/02/2023] Open
Abstract
This study was designed to assess the significance of interleukin-2 receptor (CD25) and inteleukin-3 receptor (CD123) expression in cytogenetically normal acute myeloid leukemia (CN-AML) patients. The current study includes 80 CN-AML (≤ 60 years) before the start of therapy. Blast cells expression for CD25 and CD123 were identified by flowcytometry in fresh bone marrow samples. CD25+/CD123-; CD25-/CD123+. CD25+/CD123+, CD25-/CD123- expression were as follow: 10/80 (12.5%); 18/80 (22.5%); 17/80; (21.25%), 35/80 (43.5%) respectively. The total CD25 expression was detected in 27/80 (33.75%), and CD123 expression was detected in 35/80 (43.75%%). CN-AML patients showed CD25+/CD123+ co-expression had the lowest induction remission rate and the shortest overall survival as compared to those lack co-expressions (P <0.01; P = 0.023 respectively). Also, there is strong positive association between CD25+/CD123+ co-expression and FLT3 mutations (P<0.001) and negative one with NPM1 mutation (P<0.001). In conclusion: CD25+/CD123+ co-expression in CN-AML patients define a subgroup of patients with adverse outcome. Identification of CD25/CD123 expression in CN-AML patents at diagnosis could be included in risk stratification. There is strong association between CD25+/CD123+ positive expression and FLT3 mutations.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center (MUOC), Mansoura University, Mansoura, Egypt
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University Egypt Egypt
- Corresponding author.
| | - Emaad Azmy
- Clinical Hematology Unit, Mansoura University Oncology Center (MUOC), Mansoura University, Mansoura, Egypt
| | - Lamiaa Ibrahim
- Clinical Hematology Unit, Mansoura University Oncology Center (MUOC), Mansoura University, Mansoura, Egypt
| | - Mohamed Sabry
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center (MUOC), Mansoura University, Mansoura, Egypt
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University Egypt Egypt
| | - Mohamed El Agdar
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center (MUOC), Mansoura University, Mansoura, Egypt
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University Egypt Egypt
| |
Collapse
|
56
|
Song M, Wang H, Ye Q. Increased circulating vascular endothelial growth factor in acute myeloid leukemia patients: a systematic review and meta-analysis. Syst Rev 2020; 9:103. [PMID: 32375879 PMCID: PMC7201671 DOI: 10.1186/s13643-020-01368-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is one of the angiogenesis regulators, which plays an important role in tumor angiogenesis and tumor progression. Current studies have found that VEGF plays an important role in hematologic diseases including acute myeloid leukemia (AML). However, the circulating levels of VEGF in AML were still controversial among published studies. METHODS Three databases including PubMed, EMBASE, and Cochrane Library databases were searched up to February 2020. All articles included in the meta-analysis met our inclusion and exclusion criteria. Studies will be screened and data extracted by two independent investigators. The Newcastle-Ottawa Scale (NOS) and the Risk of Bias In Non-randomized Studies of Interventions (ROBINS-I) tool were applied to evaluate the quality of the included studies. A random-effects model was applied to pool the standardized mean difference (SMD). Heterogeneity test was performed by the Q statistic and quantified using I2. All statistical analysis was conducted in Stata 12.0 software. RESULTS Fourteen case-control studies were finally included in this systematic review and meta-analysis. Heterogeneity was high in our included studies (I2 = 91.1%, P < 0.001). Sensitivity analysis showed no significant change when any one study was excluded using random-effect methods (P > 0.05). Egger's linear regression test showed that no publication bias existed (P > 0.05). Patients with AML, mainly those newly diagnosed and untreated, have higher VEGF levels (SMD = 0.85, 95% CI 0.28-1.42). Moreover, AML patients in n ≥ 40 group, plasma group, Asia and Africa group, and age ≥ 45 group had higher circulating VEGF levels (all P < 0.05). CONCLUSIONS Compared to healthy controls, our meta-analysis shows a significantly higher level of circulating VEGF in AML patients, and it is associated with sample size, sample type, region, and age.
Collapse
Affiliation(s)
- Mingzhu Song
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Huiping Wang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Qianling Ye
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China.
| |
Collapse
|
57
|
CD40 is Positively Correlated with the Expression of Nucleophosmin in Cisplatin-Resistant Bladder Cancer. JOURNAL OF ONCOLOGY 2020; 2020:3676751. [PMID: 32411234 PMCID: PMC7204159 DOI: 10.1155/2020/3676751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/19/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022]
Abstract
Objective To verify and evaluate the value of CD40 as a noninvasive biomarker of cisplatin-resistant bladder cancer, we studied the expression of CD40 and the correlation between nucleophosmin (NPM1) and CD40 in cisplatin-resistant bladder cancer. Methods Three cisplatin-resistant bladder cancer cell lines (T24/0.8DDP, BIU87/0.3DDP, and PUMC-91/0.6DDP) were studied, and lentivirus was used to silence NPM1 expression. The expression of CD40 and NPM1 in three NPM1 silencing bladder cancer cell lines were detected by fluorescence microscopy and Western Blot. The effects and proteomic bioinformatics of NPM1 gene knockout on cisplatin-resistant bladder cancer cells were analyzed by liquid chromatography-mass spectrometry (LC-MS) and gene ontology analysis (GO analysis). Results The NPM1 gene was successfully silenced in three drug-resistant bladder cancer cell lines by lentivirus infection. The knockdown efficiency was 70%. After NPM1 gene knockout, 492 differential proteins were detected by LC-MS, whose fold change was more than 1.5 (p < 0.05). A total of 57022 peptides, 54347 unique peptides, and 6686 protein groups were identified in all proteins of the tested cells (FDR < 0.01). Hierarchical clustering and principal component analysis showed that 264 functional proteins were downregulated and 228 functional proteins were upregulated in the gene silencing group compared with those of the negative controls. By GO analysis, the proteins affected by NPM1 cover a large number of proteins with biological functions, which may play an important role in the development of tumor in 492 differential proteins. The CD40 was the most significantly downregulated protein after NPM1 silencing, with a difference of 2.6-fold change in abundance. Conclusions There is a positive correlation between CD40 protein and NPM1 protein in drug-resistant bladder cancer. Because NPM1 can reflect the characteristics of bladder cancer, CD40 may be a more sensitive marker for monitoring the prognosis of bladder cancer.
Collapse
|
58
|
Qin G, Wang X, Ye S, Li Y, Chen M, Wang S, Qin T, Zhang C, Li Y, Long Q, Hu H, Shi D, Li J, Zhang K, Zhai Q, Tang Y, Kang T, Lan P, Xie F, Lu J, Deng W. NPM1 upregulates the transcription of PD-L1 and suppresses T cell activity in triple-negative breast cancer. Nat Commun 2020; 11:1669. [PMID: 32245950 PMCID: PMC7125142 DOI: 10.1038/s41467-020-15364-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 02/28/2020] [Indexed: 12/31/2022] Open
Abstract
Programmed cell death protein-1 (PD-1)/programmed cell death ligand-1 (PD-L1) interaction plays a crucial role in tumor-associated immune escape. Here, we verify that triple-negative breast cancer (TNBC) has higher PD-L1 expression than other subtypes. We then discover that nucleophosmin (NPM1) binds to PD-L1 promoter specifically in TNBC cells and activates PD-L1 transcription, thus inhibiting T cell activity in vitro and in vivo. Furthermore, we demonstrate that PARP1 suppresses PD-L1 transcription through its interaction with the nucleic acid binding domain of NPM1, which is required for the binding of NPM1 at PD-L1 promoter. Consistently, the PARP1 inhibitor olaparib elevates PD-L1 expression in TNBC and exerts a better effect with anti-PD-L1 therapy. Together, our research has revealed NPM1 as a transcription regulator of PD-L1 in TNBC, which could lead to potential therapeutic strategies to enhance the efficacy of cancer immunotherapy.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Breast/pathology
- Cell Line, Tumor
- DNA-Binding Proteins
- Disease Models, Animal
- Drug Synergism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Gene Knockdown Techniques
- Humans
- Kaplan-Meier Estimate
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Middle Aged
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleophosmin
- Phthalazines/pharmacology
- Phthalazines/therapeutic use
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors
- Poly (ADP-Ribose) Polymerase-1/metabolism
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
- Prognosis
- Promoter Regions, Genetic/genetics
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tissue Array Analysis
- Transcriptional Activation/immunology
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/mortality
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Ge Qin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Shubiao Ye
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yizhuo Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Miao Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Tao Qin
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changlin Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yixin Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Qian Long
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Huabin Hu
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dingbo Shi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jiaping Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Kai Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Qinglian Zhai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yanlai Tang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ping Lan
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fangyun Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jianjun Lu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| |
Collapse
|
59
|
Panuzzo C, Signorino E, Calabrese C, Ali MS, Petiti J, Bracco E, Cilloni D. Landscape of Tumor Suppressor Mutations in Acute Myeloid Leukemia. J Clin Med 2020; 9:jcm9030802. [PMID: 32188030 PMCID: PMC7141302 DOI: 10.3390/jcm9030802] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia is mainly characterized by a complex and dynamic genomic instability. Next-generation sequencing has significantly improved the ability of diagnostic research to molecularly characterize and stratify patients. This detailed outcome allowed the discovery of new therapeutic targets and predictive biomarkers, which led to develop novel compounds (e.g., IDH 1 and 2 inhibitors), nowadays commonly used for the treatment of adult relapsed or refractory AML. In this review we summarize the most relevant mutations affecting tumor suppressor genes that contribute to the onset and progression of AML pathology. Epigenetic modifications (TET2, IDH1 and IDH2, DNMT3A, ASXL1, WT1, EZH2), DNA repair dysregulation (TP53, NPM1), cell cycle inhibition and deficiency in differentiation (NPM1, CEBPA, TP53 and GATA2) as a consequence of somatic mutations come out as key elements in acute myeloid leukemia and may contribute to relapse and resistance to therapies. Moreover, spliceosomal machinery mutations identified in the last years, even if in a small cohort of acute myeloid leukemia patients, suggested a new opportunity to exploit therapeutically. Targeting these cellular markers will be the main challenge in the near future in an attempt to eradicate leukemia stem cells.
Collapse
Affiliation(s)
- Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Elisabetta Signorino
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Chiara Calabrese
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Muhammad Shahzad Ali
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Jessica Petiti
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Enrico Bracco
- Department of Oncology, University of Turin, 10124 Turin, Italy;
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
- Correspondence: ; Tel.: +39-011-9026610; Fax: +39-011-9038636
| |
Collapse
|
60
|
Gadewal N, Kumar R, Aher S, Gardane A, Gaur T, Varma AK, Khattry N, Hasan SK. miRNA-mRNA Profiling Reveals Prognostic Impact of SMC1A Expression in Acute Myeloid Leukemia. Oncol Res 2020; 28:321-330. [PMID: 32059753 PMCID: PMC7851519 DOI: 10.3727/096504020x15816752427321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) with NPM1 mutation is a disease driving genetic alteration with good prognosis. Although it has been suggested that NPM1 mutation induces chemosensitivity in leukemic cells, the underlying cause for the better survival of NPM1 mutated patients is still not clear. Mutant NPM1 AML has a unique microRNA and their target gene (mRNA) signature compared to wild-type NPM1. Dynamic regulation of miRNA–mRNA has been reported to influence the prognostic outcome. In the present study, in silico expression data of miRNA and mRNA in AML patients was retrieved from genome data commons, and differentially expressed miRNA and mRNA among NPM1 mutated (n = 21) and NPM1 wild-type (n = 162) cases were identified to establish a dynamic association at the molecular level. In vitro experiments using high-throughput RNA sequencing were performed on human AML cells carrying NPM1 mutated and wild-type allele. The comparison of in vitro transcriptomics data with in silico miRNA–mRNA expression network data revealed downregulation of SMC1A. On establishing miRNA–mRNA interactive pairs, it has been observed that hsa-mir-215-5p (logFC: 0.957; p = 0.0189) is involved in the downregulation of SMC1A (logFC: –0.481; p = 0.0464) in NPM1 mutated AML. We demonstrated that transient expression of NPM1 mutation upregulates miR-215-5p, which results in downregulation of SMC1A. We have also shown using a rescue experiment that neutralizing miR-215-5p reverses the effect of NPM1 mutation on SMC1A. Using the leukemic blasts from AML patients, we observed higher expression of miR-215-5p and lower expression of SMC1A in NPM1 mutated patients compared to wild-type cases. The overall survival of AML patients was significantly inferior in SMC1A high expressers compared to low expressers (20.3% vs. 58.5%, p = 0.018). The data suggest that dynamic miR-215-SMC1A regulation is potentially modulated by NPM1 mutation, which might serve as an explanation for the better outcome in NPM1 mutated AML.
Collapse
Affiliation(s)
- Nikhil Gadewal
- Bioinformatics Centre, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC)Navi MumbaiIndia
| | - Rohit Kumar
- Cell and Tumor Biology Group, Advanced Centre for Treatment, Research and Education in CancerNavi MumbaiIndia
| | - Swapnil Aher
- Cell and Tumor Biology Group, Advanced Centre for Treatment, Research and Education in CancerNavi MumbaiIndia
| | - Anagha Gardane
- Cell and Tumor Biology Group, Advanced Centre for Treatment, Research and Education in CancerNavi MumbaiIndia
| | - Tarang Gaur
- Cell and Tumor Biology Group, Advanced Centre for Treatment, Research and Education in CancerNavi MumbaiIndia
| | - Ashok K Varma
- Bioinformatics Centre, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC)Navi MumbaiIndia
| | - Navin Khattry
- Adult Hemato-lymphoid Disease Management Group, Tata Memorial HospitalMumbaiIndia
| | - Syed K Hasan
- Cell and Tumor Biology Group, Advanced Centre for Treatment, Research and Education in CancerNavi MumbaiIndia
| |
Collapse
|
61
|
Chen Y, Hu J. Nucleophosmin1 (NPM1) abnormality in hematologic malignancies, and therapeutic targeting of mutant NPM1 in acute myeloid leukemia. Ther Adv Hematol 2020; 11:2040620719899818. [PMID: 32071709 PMCID: PMC6997955 DOI: 10.1177/2040620719899818] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/18/2019] [Indexed: 01/07/2023] Open
Abstract
Nucleophosmin (NPM1) is an abundant nucleolar protein that is
implicated in a variety of biological processes and in the pathogenesis of
several human malignancies. For hematologic malignancies, approximately
one-third of anaplastic large-cell non-Hodgkin’s lymphomas were found to express
a fusion between NPM1 and the catalytic domain of anaplastic
lymphoma receptor tyrosine kinase. About 50–60% of acute myeloid leukemia
patients with normal karyotype carry NPM1 mutations, which are
characterized by cytoplasmic dislocation of the NPM1 protein.
Nevertheless, NPM1 is overexpressed in various hematologic and
solid tumor malignancies. NPM1 overexpression is considered a
prognostic marker of recurrence and progression of cancer. Thus,
NPM1 abnormalities play a critical role in several types of
hematologic malignancies. This has led to intense interest in the development of
an NPM1 targeting strategy for cancer therapy. The aim of this
review is to summarize present knowledge on NPM1 origin,
pathogenesis, and therapeutic interventions in hematologic malignancies.
Collapse
Affiliation(s)
- Yingyu Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Fuzhou, Fujian 350001, China
| | - Jianda Hu
- Department of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
62
|
Nucleophosmin mutations confer an independent favorable prognostic impact in 869 pediatric patients with acute myeloid leukemia. Blood Cancer J 2020; 10:1. [PMID: 31915364 PMCID: PMC6949268 DOI: 10.1038/s41408-019-0268-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/17/2019] [Accepted: 10/29/2019] [Indexed: 12/28/2022] Open
Abstract
Studies on the clinical significance of Nucleophosmin (NPM1) mutations in pediatric AML in a large cohort are lacking. Moreover, the prognosis of patients with co-occurring NPM1 and FLT3/ITD mutations is controversial. Here, we analyzed the impact of NPM1 mutations on prognoses of 869 pediatric AML patients from the TAGET dataset. The frequency of NPM1 mutations was 7.6%. NPM1 mutations were significantly associated with older age (P < 0.001), normal cytogenetics (P < 0.001), FLT3/ITD mutations (P < 0.001), and high complete remission induction rates (P < 0.05). Overall, NPM1-mutated patients had a significantly better 5-year EFS (P = 0.001) and OS (P = 0.016) compared to NPM1 wild-type patients, and this favorable impact was maintained even in the presence of FLT3/ITD mutations. Stem cell transplantation had no significant effect on the survival of patients with both NPM1 and FLT3/ITD mutations. Multivariate analysis revealed that NPM1 mutations were independent predictors of better outcome in terms of EFS (P = 0.004) and OS (P = 0.012). Our findings showed that NPM1 mutations confer an independent favorable prognostic impact in pediatric AML despite of FLT3/ITD mutations. In addition, pediatric AML patients with both NPM1 and FLT3/ITD mutations appear to have favorable prognoses and may not need hematopoietic stem cell transplantations.
Collapse
|
63
|
Cheng Z, Dai Y, Pang Y, Jiao Y, Liu Y, Cui L, Quan L, Qian T, Zeng T, Si C, Huang W, Chen J, Pang Y, Ye X, Shi J, Fu L. Up-regulation of DDIT4 predicts poor prognosis in acute myeloid leukaemia. J Cell Mol Med 2019; 24:1067-1075. [PMID: 31755224 PMCID: PMC6933361 DOI: 10.1111/jcmm.14831] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/23/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) inhibitor, DNA damage inducible transcript 4 (DDIT4), has inducible expression in response to various cellular stresses. In multiple malignancies, studies have shown that DDIT4 participates in tumorigenesis and impacts patient survival. We aimed to study the prognostic value of DDIT4 in acute myeloid leukaemia (AML), which is currently unclear. Firstly, The Cancer Genome Atlas was screened for AML patients with complete clinical characteristics and DDIT4 expression data. A total of 155 patients were included and stratified according to the treatment modality and the median DDIT4 expression levels. High DDIT4 expressers had shorter overall survival (OS) and event‐free survival (EFS) than the low expressers among the chemotherapy‐only group (all P < .001); EFS and OS were similar in the high and low DDIT4 expressers of the allogeneic haematopoietic stem cell transplantation (allo‐HSCT) group. Furthermore, in the DDIT4high group, patients treated with allo‐HSCT had longer EFS and OS than those who received chemotherapy alone (all P < .01). In the DDIT4low group, OS and EFS were similar in different treatment groups. Secondly, we analysed two other cytogenetically normal AML (CN‐AML) cohorts derived from the Gene Expression Omnibus database, which confirmed that high DDIT4 expression was associated with poorer survival. Gene Ontology (GO) enrichment analysis showed that the genes related to DDIT4 expression were mainly concentrated in the acute and chronic myeloid leukaemia signalling pathways. Collectively, our study indicates that high DDIT4 expression may serve as a poor prognostic factor for AML, but its prognostic effects could be outweighed by allo‐HSCT.
Collapse
Affiliation(s)
- Zhiheng Cheng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Yifeng Dai
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yifan Pang
- Department of Medicine, William Beaumont Hospital, Royal Oak, MI, USA
| | - Yang Jiao
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Liang Quan
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tiansheng Zeng
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Chaozeng Si
- Department of Operations and Information Management, China-Japan Friendship Hospital, Beijing, China
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinghong Chen
- State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Pang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu Ye
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinlong Shi
- Department of Biomedical Engineering, Chinese PLA General Hospital, Beijing, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
64
|
The Value of FLT3, NPM1 and DNMT3A Gene Mutation Analysis in Acute Myeloid Leukemia Diagnosis. REV ROMANA MED LAB 2019. [DOI: 10.2478/rrlm-2019-0024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
65
|
Liang P, Hu R, Liu Z, Miao M, Jiang H, Li C. NAT10 upregulation indicates a poor prognosis in acute myeloid leukemia. Curr Probl Cancer 2019; 44:100491. [PMID: 31279531 DOI: 10.1016/j.currproblcancer.2019.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 06/23/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND N-acetyltransferase 10 (NAT10) is considered as an oncogene in many tumors. This study investigated the NAT10 expression in Chinese acute myeloid leukemia (AML) patients and evaluated the predictive significance of NAT10 with a single-center retrospective study. METHODS The Oncomine was used to analyze NAT10 expression in AML. We also collected bone marrow samples of 48 newly diagnosed AML patients and 20 benign individuals in our center. NAT10 mRNA expression levels were detected by real-time qPCR. Clinical data was obtained from inpatient medical records. RESULTS Two microarrays in Oncomine showed that NAT10 was upregulated in AML. Our data revealed that AML patients had higher NAT10 expression levels than the normal controls (P < 0.01). NPM1-mutant patients had higher NAT10 mRNA levels than NPM1-wt patients. NAT10 expression level was higher in nonremission group than in overall remission group (P < 0.05). High NAT10 expression indicated a poor progression-free survival and overall survival. CONCLUSIONS The results support NAT10 as a potential prognostic and therapeutic biomarker for AML.
Collapse
Affiliation(s)
- Peiqi Liang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rong Hu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Zhuogang Liu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Miao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huinan Jiang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chuan Li
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
66
|
La Manna S, Scognamiglio PL, Roviello V, Borbone F, Florio D, Di Natale C, Bigi A, Cecchi C, Cascella R, Giannini C, Sibillano T, Novellino E, Marasco D. The acute myeloid leukemia-associated Nucleophosmin 1 gene mutations dictate amyloidogenicity of the C-terminal domain. FEBS J 2019; 286:2311-2328. [PMID: 30921500 DOI: 10.1111/febs.14815] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/29/2019] [Accepted: 03/21/2019] [Indexed: 01/04/2023]
Abstract
Nucleophosmin 1 (NPM1) is a nucleus-cytoplasm shuttling protein ubiquitously expressed and highly conserved. It is involved in many cellular processes and its gene is mutated in ~ 50-60% of Acute Myeloid Leukemia (AML) patients. These mutations cause its cytoplasmic mislocation and accumulation (referred to as NPM1c+) and open the door to rational targeted therapy for AML diseases with mutated NPM1. Currently, there is limited knowledge on the mechanism of action of NPM1c+ and on structural determinants of the leukemogenic potential of AML mutations. Numerous previous studies outlined an unexpected amyloid-like aggregation tendency of several regions located in the C-terminal domain that, in wild-type form, fold as a three-helical-bundle. Here, using a combination of different techniques including Thioflavin T fluorescence, congo red absorbance, CD spectroscopy, Scanning Electron Microscopy (SEM) and wide-angle X-ray scattering on a series of peptides bearing mutations, we evidence that the amyloidogenicity of NPM1 mutants is directly linked to AML. Noticeably, AML point mutations strongly affect the amyloid cytotoxic effects in neuroblastoma cells and the morphologies of deriving fibrils. This study paves the way to deepen our understanding of AML-associated NPM1 mutants, and could help to break new ground for the identification of novel drugs targeting NPM1c+ for treatment of AML.
Collapse
Affiliation(s)
- Sara La Manna
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| | - Pasqualina Liana Scognamiglio
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| | - Valentina Roviello
- Analytical Chemistry for the Environment and CeSMA (Advanced Metrologic Service Center), University of Naples "Federico II", Italy
| | - Fabio Borbone
- Department of Chemical Sciences, University of Naples "Federico II", Italy
| | - Daniele Florio
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| | - Concetta Di Natale
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| | - Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Italy
| | - Cinzia Giannini
- Institute of Crystallography (IC), National Research Council, Bari, Italy
| | - Teresa Sibillano
- Institute of Crystallography (IC), National Research Council, Bari, Italy
| | - Ettore Novellino
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| | - Daniela Marasco
- Department of Pharmacy, CIRPEB: Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II", Italy
| |
Collapse
|
67
|
CDC37 as a novel target for the treatment of NPM1-ALK expressing anaplastic large cell lymphomas. Blood Cancer J 2019; 9:14. [PMID: 30696805 PMCID: PMC6351628 DOI: 10.1038/s41408-019-0171-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/30/2018] [Accepted: 01/07/2019] [Indexed: 11/09/2022] Open
|
68
|
Minimal/Measurable Residual Disease Monitoring in NPM1-Mutated Acute Myeloid Leukemia: A Clinical Viewpoint and Perspectives. Int J Mol Sci 2018; 19:ijms19113492. [PMID: 30404199 PMCID: PMC6274702 DOI: 10.3390/ijms19113492] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) with NPM1 gene mutations is currently recognized as a distinct entity, due to its unique biological and clinical features. We summarize here the results of published studies investigating the clinical application of minimal/measurable residual disease (MRD) in patients with NPM1-mutated AML, receiving either intensive chemotherapy or hematopoietic stem cell transplantation. Several clinical trials have so far demonstrated a significant independent prognostic impact of molecular MRD monitoring in NPM1-mutated AML and, accordingly, the Consensus Document from the European Leukemia Net MRD Working Party has recently recommended that NPM1-mutated AML patients have MRD assessment at informative clinical timepoints during treatment and follow-up. However, several controversies remain, mainly with regard to the most clinically significant timepoints and the MRD thresholds to be considered, but also with respect to the optimal source to be analyzed, namely bone marrow or peripheral blood samples, and the correlation of MRD with other known prognostic indicators. Moreover, we discuss potential advantages, as well as drawbacks, of newer molecular technologies such as digital droplet PCR and next-generation sequencing in comparison to conventional RQ-PCR to quantify NPM1-mutated MRD. In conclusion, further prospective clinical trials are warranted to standardize MRD monitoring strategies and to optimize MRD-guided therapeutic interventions in NPM1-mutated AML patients.
Collapse
|
69
|
Khan I, Halasi M, Patel A, Schultz R, Kalakota N, Chen YH, Aardsma N, Liu L, Crispino JD, Mahmud N, Frankfurt O, Gartel AL. FOXM1 contributes to treatment failure in acute myeloid leukemia. JCI Insight 2018; 3:121583. [PMID: 30089730 PMCID: PMC6129129 DOI: 10.1172/jci.insight.121583] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) patients with NPM1 mutations demonstrate a superior response to standard chemotherapy treatment. Our previous work has shown that these favorable outcomes are linked to the cytoplasmic relocalization and inactivation of FOXM1 driven by mutated NPM1. Here, we went on to confirm the important role of FOXM1 in increased chemoresistance in AML. A multiinstitution retrospective study was conducted to link FOXM1 expression to clinical outcomes in AML. We establish nuclear FOXM1 as an independent clinical predictor of chemotherapeutic resistance in intermediate-risk AML in a multivariate analysis incorporating standard clinicopathologic risk factors. Using colony assays, we show a dramatic decrease in colony size and numbers in AML cell lines with knockdown of FOXM1, suggesting an important role for FOXM1 in the clonogenic activity of AML cells. In order to further prove a potential role for FOXM1 in AML chemoresistance, we induced an FLT3-ITD-driven myeloid neoplasm in a FOXM1-overexpressing transgenic mouse model and demonstrated significantly higher residual disease after standard chemotherapy. This suggests that constitutive overexpression of FOXM1 in this model induces chemoresistance. Finally, we performed proof-of-principle experiments using a currently approved proteasome inhibitor, ixazomib, to target FOXM1 and demonstrated a therapeutic response in AML patient samples and animal models of AML that correlates with the suppression of FOXM1 and its transcriptional targets. Addition of low doses of ixazomib increases sensitization of AML cells to chemotherapy backbone drugs cytarabine and the hypomethylator 5-azacitidine. Our results underscore the importance of FOXM1 in AML progression and treatment, and they suggest that targeting it may have therapeutic benefit in combination with standard AML therapies.
Collapse
Affiliation(s)
- Irum Khan
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Marianna Halasi
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | | | - Rachael Schultz
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Nandini Kalakota
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Nathan Aardsma
- Department of Pathology, University of Illinois, Chicago, Illinois, USA
| | - Li Liu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois, Chicago, Illinois, USA
| | | | - Nadim Mahmud
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | | | - Andrei L Gartel
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
70
|
Duployez N, Chebrek L, Helevaut N, Fournier E, Bemba M, Caillault A, Geffroy S, Preudhomme C. A novel type of NPM1 mutation characterized by multiple internal tandem repeats in a case of cytogenetically normal acute myeloid leukemia. Haematologica 2018; 103:e575-e577. [PMID: 29903763 DOI: 10.3324/haematol.2018.190959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Nicolas Duployez
- CHU Lille, Laboratory of Hematology .,Univ. Lille, INSERM, UMR-S 1172
| | | | - Nathalie Helevaut
- CHU Lille, Laboratory of Hematology.,Univ. Lille, INSERM, UMR-S 1172
| | - Elise Fournier
- Univ. Lille, INSERM, UMR-S 1172.,CH Dunkerque, Laboratory of Hematology, F-59385
| | - Maxime Bemba
- CH Dunkerque, Hematology Department, F-59385, France
| | | | - Sandrine Geffroy
- CHU Lille, Laboratory of Hematology.,Univ. Lille, INSERM, UMR-S 1172
| | - Claude Preudhomme
- CHU Lille, Laboratory of Hematology.,Univ. Lille, INSERM, UMR-S 1172
| |
Collapse
|