1
|
Borcherding DC, Amin NV, He K, Zhang X, Lyu Y, Dehner C, Bhatia H, Gothra A, Daud L, Ruminski P, Pratilas CA, Pollard K, Sundby T, Widemann BC, Hirbe AC. MEK Inhibition Synergizes with TYK2 Inhibitors in NF1-Associated Malignant Peripheral Nerve Sheath Tumors. Clin Cancer Res 2023; 29:1592-1604. [PMID: 36799629 PMCID: PMC10102849 DOI: 10.1158/1078-0432.ccr-22-3722] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/23/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas with limited treatment options and poor survival rates. About half of MPNST cases are associated with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Overexpression of TYK2 occurs in the majority of MPNST, implicating TYK2 as a therapeutic target. EXPERIMENTAL DESIGN The effects of pharmacologic TYK2 inhibition on MPNST cell proliferation and survival were examined using IncuCyte live cell assays in vitro, and downstream actions were analyzed using RNA-sequencing (RNA-seq), qPCR arrays, and validation of protein changes with the WES automated Western system. Inhibition of TYK2 alone and in combination with MEK inhibition was evaluated in vivo using both murine and human MPNST cell lines, as well as MPNST PDX. RESULTS Pharmacologic inhibition of TYK2 dose-dependently decreased proliferation and induced apoptosis over time. RNA-seq pathway analysis on TYK2 inhibitor-treated MPNST demonstrated decreased expression of cell cycle, mitotic, and glycolysis pathways. TYK2 inhibition resulted in upregulation of the MEK/ERK pathway gene expression, by both RNA-seq and qPCR array, as well as increased pERK1/2 levels by the WES Western system. The compensatory response was tested with dual treatment with TYK2 and MEK inhibitors, which synergistically decreased proliferation and increased apoptosis in vitro. Finally, combination therapy was shown to inhibit growth of MPNST in multiple in vivo models. CONCLUSIONS These data provide the preclinical rationale for the development of a phase I clinical trial of deucravacitinib and mirdametinib in NF1-assosciated MPNST.
Collapse
Affiliation(s)
- Dana C. Borcherding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Neha V. Amin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin He
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaochun Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yang Lyu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Carina Dehner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Himanshi Bhatia
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Angad Gothra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Layla Daud
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Ruminski
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Christine A. Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Taylor Sundby
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Angela C. Hirbe
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
2
|
Shah K, Al Ashiri L, Nasimian A, Ahmed M, Kazi JU. Venetoclax-Resistant T-ALL Cells Display Distinct Cancer Stem Cell Signatures and Enrichment of Cytokine Signaling. Int J Mol Sci 2023; 24:ijms24055004. [PMID: 36902436 PMCID: PMC10003524 DOI: 10.3390/ijms24055004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Therapy resistance remains one of the major challenges for cancer treatment that largely limits treatment benefits and patient survival. The underlying mechanisms that lead to therapy resistance are highly complicated because of the specificity to the cancer subtype and therapy. The expression of the anti-apoptotic protein BCL2 has been shown to be deregulated in T-cell acute lymphoblastic leukemia (T-ALL), where different T-ALL cells display a differential response to the BCL2-specific inhibitor venetoclax. In this study, we observed that the expression of anti-apoptotic BCL2 family genes, such as BCL2, BCL2L1, and MCL1, is highly varied in T-ALL patients, and inhibitors targeting proteins coded by these genes display differential responses in T-ALL cell lines. Three T-ALL cell lines (ALL-SIL, MOLT-16, and LOUCY) were highly sensitive to BCL2 inhibition within a panel of cell lines tested. These cell lines displayed differential BCL2 and BCL2L1 expression. Prolonged exposure to venetoclax led to the development of resistance to it in all three sensitive cell lines. To understand how cells developed venetoclax resistance, we monitored the expression of BCL2, BCL2L1, and MCL1 over the treatment period and compared gene expression between resistant cells and parental sensitive cells. We observed a different trend of regulation in terms of BCL2 family gene expression and global gene expression profile including genes reported to be expressed in cancer stem cells. Gene set enrichment analysis (GSEA) showed enrichment of cytokine signaling in all three cell lines which was supported by the phospho-kinase array where STAT5 phosphorylation was found to be elevated in resistant cells. Collectively, our data suggest that venetoclax resistance can be mediated through the enrichment of distinct gene signatures and cytokine signaling pathways.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Lina Al Ashiri
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Ahmad Nasimian
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Mehreen Ahmed
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 22381 Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 22184 Lund, Sweden
- Correspondence:
| |
Collapse
|
3
|
Heat-Shock Proteins in Leukemia and Lymphoma: Multitargets for Innovative Therapeutic Approaches. Cancers (Basel) 2023; 15:cancers15030984. [PMID: 36765939 PMCID: PMC9913431 DOI: 10.3390/cancers15030984] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Heat-shock proteins (HSPs) are powerful chaperones that provide support for cellular functions under stress conditions but also for the homeostasis of basic cellular machinery. All cancer cells strongly rely on HSPs, as they must continuously adapt to internal but also microenvironmental stresses to survive. In solid tumors, HSPs have been described as helping to correct the folding of misfolded proteins, sustain oncogenic pathways, and prevent apoptosis. Leukemias and lymphomas also overexpress HSPs, which are frequently associated with resistance to therapy. HSPs have therefore been proposed as new therapeutic targets. Given the specific biology of hematological malignancies, it is essential to revise their role in this field, providing a more adaptable and comprehensive picture that would help design future clinical trials. To that end, this review will describe the different pathways and functions regulated by HSP27, HSP70, HSP90, and, not least, HSP110 in leukemias and lymphomas.
Collapse
|
4
|
Mustafa AHM, Krämer OH. Pharmacological Modulation of the Crosstalk between Aberrant Janus Kinase Signaling and Epigenetic Modifiers of the Histone Deacetylase Family to Treat Cancer. Pharmacol Rev 2023; 75:35-61. [PMID: 36752816 DOI: 10.1124/pharmrev.122.000612] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/08/2022] [Accepted: 08/15/2022] [Indexed: 12/13/2022] Open
Abstract
Hyperactivated Janus kinase (JAK) signaling is an appreciated drug target in human cancers. Numerous mutant JAK molecules as well as inherent and acquired drug resistance mechanisms limit the efficacy of JAK inhibitors (JAKi). There is accumulating evidence that epigenetic mechanisms control JAK-dependent signaling cascades. Like JAKs, epigenetic modifiers of the histone deacetylase (HDAC) family regulate the growth and development of cells and are often dysregulated in cancer cells. The notion that inhibitors of histone deacetylases (HDACi) abrogate oncogenic JAK-dependent signaling cascades illustrates an intricate crosstalk between JAKs and HDACs. Here, we summarize how structurally divergent, broad-acting as well as isoenzyme-specific HDACi, hybrid fusion pharmacophores containing JAKi and HDACi, and proteolysis targeting chimeras for JAKs inactivate the four JAK proteins JAK1, JAK2, JAK3, and tyrosine kinase-2. These agents suppress aberrant JAK activity through specific transcription-dependent processes and mechanisms that alter the phosphorylation and stability of JAKs. Pharmacological inhibition of HDACs abrogates allosteric activation of JAKs, overcomes limitations of ATP-competitive type 1 and type 2 JAKi, and interacts favorably with JAKi. Since such findings were collected in cultured cells, experimental animals, and cancer patients, we condense preclinical and translational relevance. We also discuss how future research on acetylation-dependent mechanisms that regulate JAKs might allow the rational design of improved treatments for cancer patients. SIGNIFICANCE STATEMENT: Reversible lysine-ɛ-N acetylation and deacetylation cycles control phosphorylation-dependent Janus kinase-signal transducer and activator of transcription signaling. The intricate crosstalk between these fundamental molecular mechanisms provides opportunities for pharmacological intervention strategies with modern small molecule inhibitors. This could help patients suffering from cancer.
Collapse
Affiliation(s)
- Al-Hassan M Mustafa
- Department of Toxicology, University Medical Center, Mainz, Germany (A.-H.M.M., O.H.K.) and Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt (A.-H.M.M.)
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany (A.-H.M.M., O.H.K.) and Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt (A.-H.M.M.)
| |
Collapse
|
5
|
Thomas X. T-cell acute lymphoblastic leukemia: promising experimental drugs in clinical development. Expert Opin Investig Drugs 2023; 32:37-52. [PMID: 36541671 DOI: 10.1080/13543784.2023.2161361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Despite advances in treatment approaches in acute lymphoblastic leukemia (ALL), the prognosis of adults with newly diagnosed T-ALL remains poor, as well as that of adults and children with relapsed disease. Novel targeted therapies are therefore needed. AREAS COVERED This review summarizes promising emerging strategies for the treatment of T-ALL. EXPERT OPINION The recent molecular characterization of T-ALL has led to the identification of new therapeutic targets. Small-molecules inhibitors and other targeted therapies have therefore been recently developed and are currently under clinical investigations. Similarly, first studies involving monoclonal antibodies and chimeric antigen receptor (CAR) T cells have shown encouraging results. Improvement of outcome with these novel approaches, eventually combined with current standard chemotherapy, is therefore expected in a near future in T-ALL.
Collapse
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Department of Clinical Hematology, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
6
|
Therapeutic Targeting of MERTK and BCL-2 in T-Cell and Early T-Precursor Acute Lymphoblastic Leukemia. Cancers (Basel) 2022; 14:cancers14246142. [PMID: 36551626 PMCID: PMC9776749 DOI: 10.3390/cancers14246142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) accounts for 15% of childhood ALL. The early T-precursor (ETP-ALL) subset is characterized by an immature T-cell phenotype, chemoresistance, and high rates of induction failure. MERTK receptor tyrosine kinase is ectopically expressed in half of T-ALLs, particularly those with an immature T-cell phenotype, suggesting a role in ETP-ALL. The anti-apoptotic protein B-cell lymphoma-2 (BCL-2) is essential for ETP-ALL cell survival. Here, we show that MERTK and BCL-2 mRNA and protein are preferentially expressed in ETP-ALL patient samples. The dual MERTK/FLT3 inhibitor MRX-2843 decreased MERTK activation and downstream signaling, inhibited cell expansion, and induced cell death in ETP-ALL cell lines. Further, 54% (21/39) of primary T-ALL patient samples were sensitive to MERTK inhibition. Treatment with MRX-2843 significantly reduced leukemia burden and prolonged survival in cell-line-derived T-ALL and ETP-ALL xenograft models. In a patient-derived ETP-ALL xenograft model, treatment with MRX-2843 markedly reduced peripheral blood leukemia and spleen weight compared to vehicle-treated mice and prolonged survival. MRX-2843 also synergized with venetoclax to provide enhanced anti-leukemia activity in ETP-ALL cell cultures, with a dose ratio of 1:20 MRX-2843:venetoclax providing optimal synergy. These data demonstrate the therapeutic potential of MRX-2843 in patients with T-ALL and provide rationale for clinical development. MRX-2843 monotherapy is currently being tested in patients with relapsed leukemia (NCT04872478). Further, our data indicate that combined MERTK and BCL-2 inhibition may be particularly effective for treatment of ETP-ALL.
Collapse
|
7
|
The combination of ruxolitinib and Bcl-2/Mcl-1 inhibitors has a synergistic effect on leukemic cells carrying a SPAG9::JAK2 fusion. Cancer Gene Ther 2022; 29:1930-1938. [PMID: 35879405 DOI: 10.1038/s41417-022-00511-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/26/2022] [Accepted: 07/13/2022] [Indexed: 01/25/2023]
Abstract
JAK2 rearrangements can occur in Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL). Here, we performed functional analysis of the SPAG9::JAK2 fusion, which was identified in a pediatric patient with Ph-like ALL, to establish molecular targeted therapy. Ba/F3 cells expressing SPAG9::JAK2 generated by retroviral transduction (Ba/F3-SPAG9-JAK2), proliferated in the absence of IL-3, and exhibited constitutive phosphorylation of the tyrosine residues in the JAK2 kinase domain of the fusion protein and STAT3/STAT5. Mutation of tyrosine residues in the JAK2 kinase domain (SPAG9::JAK2 mut) abolished IL-3 independence, but had no influence on STAT3/STAT5 phosphorylation levels. Gene expression analysis revealed that Stat1 was significantly upregulated in Ba/F3-SPAG9-JAK2 cells. STAT1 was also phosphorylated in Ba/F3-SPAG9-JAK2 but not SPAG9-JAK2 mut cells, suggesting that STAT1 is key for SPAG9::JAK2-mediated cell proliferation. Consistently, STAT1 induced expression of the anti-apoptotic proteins, BCL-2 and MCL-1, as did SPAG9::JAK2, but not SPAG9::JAK2 mut. Ruxolitinib abrogated Ba/F3-SPAG9-JAK2-mediated proliferation in vitro, but was insufficient in vivo. Venetoclax (a BCL-2 inhibitor) or AZD5991 (an MCL-1 inhibitor) enhanced the effects of ruxolitinib on Ba/F3-SPAG9-JAK2 in vitro. These findings suggest that activation of the JAK2-STAT1-BCL-2/MCL-1 axis contributes to SPAG9::JAK2-related aberrant growth promotion. BCL-2 or MCL-1 inhibition is a potential therapeutic option for B-ALL with SPAG9::JAK2 fusion.
Collapse
|
8
|
Ge J, Mu S, Xiao E, Tian G, Tao L, Li D. Expression, oncological and immunological characterizations of BZW1/2 in pancreatic adenocarcinoma. Front Genet 2022; 13:1002673. [PMID: 36267402 PMCID: PMC9576853 DOI: 10.3389/fgene.2022.1002673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Despite the progress in early diagnosis and treatment, prognosis of pancreatic adenocarcinoma (PAAD) is still poor. Basic leucine zipper and W2 domain-containing protein 1 (BZW1) and protein 2 (BZW2) are attached to the basic leucine zipper (bZIP) superfamily. Recently, BZW1 was identified as an important role in glycolysis of PAAD. However, the comprehensive reports about BZW1/2 in PAAD are not sufficient. Methods: RNA-seq data in the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were retrospectively analyzed. We explored the expression of BZW1/2 in PAAD tissues and the associations between BZW1/2 and prognosis. In addition, the potential roles of BZW1/2 in tumor microenvironment (TME) of PAAD were analyzed. Finally, clinicopathological data of 49 patients with PAAD in our institution were collected. Immunohistochemistry was used to determine the expression of BZW1/2 in PAAD samples. Results: BZW1 and BZW2 were upregulated in PAAD tissues compared to normal tissues (p < 0.05). The expression of BZW1/2 were not significantly correlated with gender, grade and stage of PAAD (p > 0.05). High expression of BZW2 was an independent predictor for poor prognosis of PAAD (HR 1.834, 95%CI 1.303–2.581, p = 0.001). And a nomogram to predict overall survival (OS) of PAAD was established with a C-index of 0.685. BZW1 and BZW2 expression were positively associated with T cell mediated immune response to tumor cell and Th2 cells in xCell database. Tumor Immune Single-Cell Hub (TISCH) analyses indicated that BZW1 and BZW2 were mainly expressed in B cells and malignant cells. External cohort furtherly validated that high expression of BZW1 and BZW2 were predictors for poor prognosis of PAAD. Conclusion: We found that BZW1 and BZW2 are highly expressed in malignant cells and B cells in the TME of PAAD. BZW2 is an independent predictor for OS of PAAD. BZW1 and BZW2 expression are positively associated with T cell mediated immune response to tumor cell and Th2 cells in PAAD.
Collapse
Affiliation(s)
| | | | | | | | | | - Deyu Li
- *Correspondence: Lianyuan Tao, ; Deyu Li,
| |
Collapse
|
9
|
Moritsch S, Mödl B, Scharf I, Janker L, Zwolanek D, Timelthaler G, Casanova E, Sibilia M, Mohr T, Kenner L, Herndler-Brandstetter D, Gerner C, Müller M, Strobl B, Eferl R. Tyk2 is a tumor suppressor in colorectal cancer. Oncoimmunology 2022; 11:2127271. [PMID: 36185806 PMCID: PMC9519006 DOI: 10.1080/2162402x.2022.2127271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 12/04/2022] Open
Abstract
Janus kinase Tyk2 is implicated in cancer immune surveillance, but its role in solid tumors is not well defined. We used Tyk2 knockout mice (Tyk2Δ/Δ) and mice with conditional deletion of Tyk2 in hematopoietic (Tyk2ΔHem) or intestinal epithelial cells (Tyk2ΔIEC) to assess their cell type-specific functions in chemically induced colorectal cancer. All Tyk2-deficient mouse models showed a higher tumor burden after AOM-DSS treatment compared to their corresponding wild-type controls (Tyk2+/+ and Tyk2fl/fl), demonstrating tumor-suppressive functions of Tyk2 in immune cells and epithelial cancer cells. However, specific deletion of Tyk2 in hematopoietic cells or in intestinal epithelial cells was insufficient to accelerate tumor progression, while deletion in both compartments promoted carcinoma formation. RNA-seq and proteomics revealed that tumors of Tyk2Δ/Δ and Tyk2ΔIEC mice were immunoedited in different ways with downregulated and upregulated IFNγ signatures, respectively. Accordingly, the IFNγ-regulated immune checkpoint Ido1 was downregulated in Tyk2Δ/Δ and upregulated in Tyk2ΔIEC tumors, although both showed reduced CD8+ T cell infiltration. These data suggest that Tyk2Δ/Δ tumors are Ido1-independent and poorly immunoedited while Tyk2ΔIEC tumors require Ido1 for immune evasion. Our study shows that Tyk2 prevents Ido1 expression in CRC cells and promotes CRC immune surveillance in the tumor stroma. Both of these Tyk2-dependent mechanisms must work together to prevent CRC progression.
Collapse
Affiliation(s)
- Stefan Moritsch
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Bernadette Mödl
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Irene Scharf
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolomics Facility, University and Medical University of Vienna, Vienna, Austria
| | - Daniela Zwolanek
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Gerald Timelthaler
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Emilio Casanova
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Maria Sibilia
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Thomas Mohr
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| | - Lukas Kenner
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolomics Facility, University and Medical University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Robert Eferl
- Center for Cancer Research, Medical University of Vienna & Comprehensive Cancer Center, Vienna, Austria
| |
Collapse
|
10
|
Qu H, Wang M, Wang M, Liu Y, Quan C. The expression and the tumor suppressor role of CLDN6 in colon cancer. Mol Cell Biochem 2022; 477:2883-2893. [PMID: 35701678 DOI: 10.1007/s11010-022-04450-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/24/2022] [Indexed: 11/29/2022]
Abstract
As a member of the tight junction family, CLDN6 is a tumor suppressor in breast cancer, but its role in colon cancer is unknown. In this research, we aimed at revealing the function of CLDN6 in colon cancer. We found that colon cancer tissues lowly expressed CLDN6, and the expression of CLDN6 was negatively correlated with lymph node metastasis. Similarly, CLDN6 was lowly expressed in the colon cancer cell line SW1116, and overexpression of CLDN6 inhibited cell proliferation in vitro and in vivo. Consistently, the migration and invasion abilities of cells were significantly inhibited after CLDN6 overexpression. In addition, we demonstrated that CLDN6 may inhibit the migration and invasion abilities by activating the TYK2/STAT3 pathway. Therefore, our data indicated that CLDN6 acted as a tumor suppressor and had the potential to be regarded as a biomarker for the progression of colon cancer.
Collapse
Affiliation(s)
- Huinan Qu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Min Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Yuanyuan Liu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
11
|
Bodaar K, Yamagata N, Barthe A, Landrigan J, Chonghaile TN, Burns M, Stevenson KE, Devidas M, Loh ML, Hunger SP, Wood B, Silverman LB, Teachey DT, Meijerink JP, Letai A, Gutierrez A. JAK3 mutations and mitochondrial apoptosis resistance in T-cell acute lymphoblastic leukemia. Leukemia 2022; 36:1499-1507. [PMID: 35411095 PMCID: PMC9177679 DOI: 10.1038/s41375-022-01558-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 11/09/2022]
Abstract
Resistance to mitochondrial apoptosis predicts inferior treatment outcomes in patients with diverse tumor types, including T-cell acute lymphoblastic leukemia (T-ALL). However, the genetic basis for variability in this mitochondrial apoptotic phenotype is poorly understood, preventing its rational therapeutic targeting. Using BH3 profiling and exon sequencing analysis of childhood T-ALL clinical specimens, we found that mitochondrial apoptosis resistance was most strongly associated with activating mutations of JAK3. Mutant JAK3 directly repressed apoptosis in leukemia cells, because its inhibition with mechanistically distinct pharmacologic inhibitors resulted in reversal of mitochondrial apoptotic blockade. Inhibition of JAK3 led to loss of MEK, ERK and BCL2 phosphorylation, and BH3 profiling revealed that JAK3-mutant primary T-ALL patient samples were characterized by a dependence on BCL2. Treatment of JAK3-mutant T-ALL cells with the JAK3 inhibitor tofacitinib in combination with a spectrum of conventional chemotherapeutics revealed synergy with glucocorticoids, in vitro and in vivo. These findings thus provide key insights into the molecular genetics of mitochondrial apoptosis resistance in childhood T-ALL, and a compelling rationale for a clinical trial of JAK3 inhibitors in combination with glucocorticoids for patients with JAK3-mutant T-ALL.
Collapse
Affiliation(s)
- Kimberly Bodaar
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Natsuko Yamagata
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anais Barthe
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jack Landrigan
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Triona Ni Chonghaile
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.,Deparment of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Melissa Burns
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Kristen E. Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Meenakshi Devidas
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mignon L. Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, and the Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, 98105, USA
| | - Stephen P. Hunger
- Division of Oncology and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Brent Wood
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Lewis B. Silverman
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David T. Teachey
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Alejandro Gutierrez
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA. .,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Xu J, Zhang J, Mao QF, Wu J, Wang Y. The Interaction Between Autophagy and JAK/STAT3 Signaling Pathway in Tumors. Front Genet 2022; 13:880359. [PMID: 35559037 PMCID: PMC9086235 DOI: 10.3389/fgene.2022.880359] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/21/2022] [Indexed: 01/30/2023] Open
Abstract
Tumor is one of the important factors affecting human life and health in today’s world, and scientists have studied it extensively and deeply, among which autophagy and JAK/STAT3 signaling pathway are two important research directions. The JAK/STAT3 axis is a classical intracellular signaling pathway that assumes a key role in the regulation of cell proliferation, apoptosis, and vascular neogenesis, and its abnormal cell signaling and regulation are closely related to the occurrence and development of tumors. Therefore, the JAK/STAT3 pathway in tumor cells and various stromal cells in their microenvironment is often considered as an effective target for tumor therapy. Autophagy is a process that degrades cytoplasmic proteins and organelles through the lysosomal pathway. It is a fundamental metabolic mechanism for intracellular degradation. The mechanism of action of autophagy is complex and may play different roles at various stages of tumor development. Altered STAT3 expression has been found to be accompanied by the abnormal autophagy activity in many oncological studies, and the two may play a synergistic or antagonistic role in promoting or inhibiting the occurrence and development of tumors. This article reviews the recent advances in autophagy and its interaction with JAK/STAT3 signaling pathway in the pathogenesis, prevention, diagnosis, and treatment of tumors.
Collapse
Affiliation(s)
- Jiangyan Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinrong Zhang
- Department of Science and Education, Dafeng District People's Hospital, Yancheng, China
| | - Qi-Fen Mao
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuan Wang
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
13
|
Najar MA, Arefian M, Sidransky D, Gowda H, Prasad TSK, Modi PK, Chatterjee A. Tyrosine Phosphorylation Profiling Revealed the Signaling Network Characteristics of CAMKK2 in Gastric Adenocarcinoma. Front Genet 2022; 13:854764. [PMID: 35646067 PMCID: PMC9136244 DOI: 10.3389/fgene.2022.854764] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) is a serine/threonine protein kinase which functions via the calcium-triggered signaling cascade with CAMK1, CAMK4, and AMPKα as the immediate downstream substrates. CAMKK2 is reported to be overexpressed in gastric cancer; however, its signaling mechanism is poorly understood. We carried out label-free quantitative tyrosine phosphoproteomics to investigate tyrosine-mediated molecular signaling associated with CAMKK2 in gastric cancer cells. Using a high-resolution Orbitrap Fusion Tribrid Fourier-transform mass spectrometer, we identified 350 phosphotyrosine sites mapping to 157 proteins. We observed significant alterations in 81 phosphopeptides corresponding to 63 proteins upon inhibition of CAMKK2, among which 16 peptides were hyperphosphorylated corresponding to 13 proteins and 65 peptides were hypophosphorylated corresponding to 51 proteins. We report here that the inhibition of CAMKK2 leads to changes in the phosphorylation of several tyrosine kinases such as PKP2, PTK2, EPHA1, EPHA2, PRKCD, MAPK12, among others. Pathway analyses revealed that proteins are differentially phosphorylated in response to CAMKK2 inhibition involved in focal adhesions, actin cytoskeleton, axon guidance, and signaling by VEGF. The western blot analysis upon inhibition and/or silencing of CAMKK2 revealed a decrease in phosphorylation of PTK2 at Y925, c-JUN at S73, and STAT3 at Y705, which was in concordance with the mass spectrometry data. The study indicates that inhibition of CAMKK2 has an anti-oncogenic effect in gastric cells regulating phosphorylation of STAT3 through PTK2/c-JUN in gastric cancer.
Collapse
Affiliation(s)
- Mohd. Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Mohammad Arefian
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - David Sidransky
- Department of Oncology and Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Harsha Gowda
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- *Correspondence: Prashant Kumar Modi, ; Aditi Chatterjee,
| | - Aditi Chatterjee
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- *Correspondence: Prashant Kumar Modi, ; Aditi Chatterjee,
| |
Collapse
|
14
|
Regulation of activated T cell survival in rheumatic autoimmune diseases. Nat Rev Rheumatol 2022; 18:232-244. [PMID: 35075294 DOI: 10.1038/s41584-021-00741-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/29/2022]
Abstract
Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.
Collapse
|
15
|
Erdogan F, Qadree AK, Radu TB, Orlova A, de Araujo ED, Israelian J, Valent P, Mustjoki SM, Herling M, Moriggl R, Gunning PT. Structural and mutational analysis of member-specific STAT functions. Biochim Biophys Acta Gen Subj 2022; 1866:130058. [PMID: 34774983 DOI: 10.1016/j.bbagen.2021.130058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND The STAT family of transcription factors control gene expression in response to signals from various stimulus. They display functions in diseases ranging from autoimmunity and chronic inflammatory disease to cancer and infectious disease. SCOPE OF REVIEW This work uses an approach informed by structural data to explore how domain-specific structural variations, post-translational modifications, and the cancer genome mutational landscape dictate STAT member-specific activities. MAJOR CONCLUSIONS We illustrated the structure-function relationship of STAT proteins and highlighted their effect on member-specific activity. We correlated disease-linked STAT mutations to the structure and cancer genome mutational landscape and proposed rational drug targeting approaches of oncogenic STAT pathway addiction. GENERAL SIGNIFICANCE Hyper-activated STATs and their variants are associated with multiple diseases and are considered high value oncology targets. A full understanding of the molecular basis of member-specific STAT-mediated signaling and the strategies to selectively target them requires examination of the difference in their structures and sequences.
Collapse
Affiliation(s)
- Fettah Erdogan
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Abdul K Qadree
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Tudor B Radu
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Elvin D de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada
| | - Johan Israelian
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Satu M Mustjoki
- Hematology Research Unit, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Marco Herling
- Department of Hematology, Cellular Therapy, and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Patrick T Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada.
| |
Collapse
|
16
|
Cordo’ V, Meijer MT, Hagelaar R, de Goeij-de Haas RR, Poort VM, Henneman AA, Piersma SR, Pham TV, Oshima K, Ferrando AA, Zaman GJR, Jimenez CR, Meijerink JPP. Phosphoproteomic profiling of T cell acute lymphoblastic leukemia reveals targetable kinases and combination treatment strategies. Nat Commun 2022; 13:1048. [PMID: 35217681 PMCID: PMC8881579 DOI: 10.1038/s41467-022-28682-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/26/2022] [Indexed: 01/05/2023] Open
Abstract
Protein kinase inhibitors are amongst the most successful cancer treatments, but targetable kinases activated by genomic abnormalities are rare in T cell acute lymphoblastic leukemia. Nevertheless, kinases can be activated in the absence of genetic defects. Thus, phosphoproteomics can provide information on pathway activation and signaling networks that offer opportunities for targeted therapy. Here, we describe a mass spectrometry-based global phosphoproteomic profiling of 11 T cell acute lymphoblastic leukemia cell lines to identify targetable kinases. We report a comprehensive dataset consisting of 21,000 phosphosites on 4,896 phosphoproteins, including 217 kinases. We identify active Src-family kinases signaling as well as active cyclin-dependent kinases. We validate putative targets for therapy ex vivo and identify potential combination treatments, such as the inhibition of the INSR/IGF-1R axis to increase the sensitivity to dasatinib treatment. Ex vivo validation of selected drug combinations using patient-derived xenografts provides a proof-of-concept for phosphoproteomics-guided design of personalized treatments.
Collapse
Affiliation(s)
- Valentina Cordo’
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mariska T. Meijer
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rico Hagelaar
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Richard R. de Goeij-de Haas
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Vera M. Poort
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Alex A. Henneman
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Sander R. Piersma
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Thang V. Pham
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Koichi Oshima
- grid.239585.00000 0001 2285 2675Institute for Cancer Genetics, Columbia University Medical Center, New York, NY USA
| | - Adolfo A. Ferrando
- grid.239585.00000 0001 2285 2675Institute for Cancer Genetics, Columbia University Medical Center, New York, NY USA
| | | | - Connie R. Jimenez
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Jules P. P. Meijerink
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands ,Present Address: Acerta Pharma (member of the AstraZeneca group), Oss, The Netherlands
| |
Collapse
|
17
|
Nachmias B, Khan DH, Voisin V, Mer AS, Thomas GE, Segev N, St-Germain J, Hurren R, Gronda M, Botham A, Wang X, Maclean N, Seneviratne AK, Duong N, Xu C, Arruda A, Orouji E, Algouneh A, Hakem R, Shlush L, Minden MD, Raught B, Bader GD, Schimmer AD. IPO11 regulates the nuclear import of BZW1/2 and is necessary for AML cells and stem cells. Leukemia 2022; 36:1283-1295. [PMID: 35152270 PMCID: PMC9061300 DOI: 10.1038/s41375-022-01513-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 12/24/2022]
Abstract
AML cells are arranged in a hierarchy with stem/progenitor cells giving rise to more differentiated bulk cells. Despite the importance of stem/progenitors in the pathogenesis of AML, the determinants of the AML stem/progenitor state are not fully understood. Through a comparison of genes that are significant for growth and viability of AML cells by way of a CRISPR screen, with genes that are differentially expressed in leukemia stem cells (LSC), we identified importin 11 (IPO11) as a novel target in AML. Importin 11 (IPO11) is a member of the importin β family of proteins that mediate transport of proteins across the nuclear membrane. In AML, knockdown of IPO11 decreased growth, reduced engraftment potential of LSC, and induced differentiation. Mechanistically, we identified the transcription factors BZW1 and BZW2 as novel cargo of IPO11. We further show that BZW1/2 mediate a transcriptional signature that promotes stemness and survival of LSC. Thus, we demonstrate for the first time how specific cytoplasmic-nuclear regulation supports stem-like transcriptional signature in relapsed AML.
Collapse
|
18
|
Functional characterization of NPM1-TYK2 fusion oncogene. NPJ Precis Oncol 2022; 6:3. [PMID: 35042970 PMCID: PMC8766497 DOI: 10.1038/s41698-021-00246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 12/16/2021] [Indexed: 11/08/2022] Open
Abstract
Gene fusions are known to drive many human cancers. Therefore, the functional characterization of newly discovered fusions is critical to understanding the oncobiology of these tumors and to enable therapeutic development. NPM1–TYK2 is a novel fusion identified in CD30 + lymphoproliferative disorders, and here we present the functional evaluation of this fusion gene as an oncogene. The chimeric protein consists of the amino-terminus of nucleophosmin 1 (NPM1) and the carboxyl-terminus of tyrosine kinase 2 (TYK2), including the kinase domain. Using in vitro lymphoid cell transformation assays and in vivo tumorigenic xenograft models we present direct evidence that the fusion gene is an oncogene. NPM1 fusion partner provides the critical homodimerization needed for the fusion kinase constitutive activation and downstream signaling that are responsible for cell transformation. As a result, our studies identify NPM1–TYK2 as a novel fusion oncogene and suggest that inhibition of fusion homodimerization could be a precision therapeutic approach in cutaneous T-cell lymphoma patients expressing this chimera.
Collapse
|
19
|
Exploring the oncogenic and therapeutic target potential of the MYB-TYK2 fusion gene in B-cell acute lymphoblastic leukemia. Cancer Gene Ther 2022; 29:1140-1152. [PMID: 35022522 DOI: 10.1038/s41417-021-00421-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/01/2021] [Accepted: 12/15/2021] [Indexed: 11/08/2022]
Abstract
TYK2-rearrangements have recently been identified in high-risk acute lymphoblastic leukemia (HR-ALL) cases and are associated with poor outcome. Current understanding of the leukemogenic potential and therapeutic targetability of activating TYK2 alterations in the ALL setting is unclear, thus further investigations are warranted. Consequently, we developed in vitro, and for the first time, in vivo models of B-cell ALL from a patient harboring the MYB-TYK2 fusion gene. These models revealed JAK/STAT signaling activation and the oncogenic potential of the MYB-TYK2 fusion gene in isolation. High throughput screening identified the HDAC inhibitor, vorinostat and the HSP90 inhibitor, tanespimycin plus the JAK inhibitor, cerdulatinib as the most effective agents against cells expressing the MYB-TYK2 alteration. Evaluation of vorinostat and cerdulatinib in pre-clinical models of MYB-TYK2-rearranged ALL demonstrated that both drugs exhibited anti-leukemic effects and reduced the disease burden in treated mice. Importantly, these findings indicate that activating TYK2 alterations can function as driver oncogenes rather than passenger or secondary events in disease development. In addition, our data provide evidence for use of vorinostat and cerdulatinib in the treatment regimens of patients with this rare yet aggressive type of high-risk ALL that warrants further investigation in the clinical setting.
Collapse
|
20
|
Murai Y, Jo U, Murai J, Fukuda S, Takebe N, Pommier Y. Schlafen 11 expression in human acute leukemia cells with gain-of-function mutations in the interferon-JAK signaling pathway. iScience 2021; 24:103173. [PMID: 34693224 PMCID: PMC8517841 DOI: 10.1016/j.isci.2021.103173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/16/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022] Open
Abstract
Schlafen11 (SLFN11) is referred to as interferon (IFN)-inducible. Based on cancer genomic databases, we identified human acute myeloid and lymphoblastic leukemia cells with gain-of-function mutations in the Janus kinase (JAK) family as exhibiting high SLFN11 expression. In these cells, the clinical JAK inhibitors cerdulatinib, ruxolitinib, and tofacitinib reduced SLFN11 expression, but IFN did not further induce SLFN11 despite phosphorylated STAT1. We provide evidence that suppression of SLFN11 by JAK inhibitors is caused by inactivation of the non-canonical IFN pathway controlled by AKT and ERK. Accordingly, the AKT and ERK inhibitors MK-2206 and SCH77284 suppressed SLFN11 expression. Both also suppressed the E26 transformation-specific (ETS)-family genes ETS-1 and FLI-1 that act as transcription factors for SLFN11. Moreover, SLFN11 expression was inhibited by the ETS inhibitor TK216. Our study reveals that SLFN11 expression is regulated via the JAK, AKT and ERK, and ETS axis. Pharmacological suppression of SLFN11 warrants future studies.
Collapse
Affiliation(s)
- Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Naoko Takebe
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Developmental Therapeutics Branch and Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
21
|
Silva A, Almeida ARM, Cachucho A, Neto JL, Demeyer S, de Matos M, Hogan T, Li Y, Meijerink J, Cools J, Grosso AR, Seddon B, Barata JT. Overexpression of wild-type IL-7Rα promotes T-cell acute lymphoblastic leukemia/lymphoma. Blood 2021; 138:1040-1052. [PMID: 33970999 PMCID: PMC8462360 DOI: 10.1182/blood.2019000553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/15/2021] [Indexed: 12/02/2022] Open
Abstract
Tight regulation of IL-7Rα expression is essential for normal T-cell development. IL-7Rα gain-of-function mutations are known drivers of T-cell acute lymphoblastic leukemia (T-ALL). Although a subset of patients with T-ALL display high IL7R messenger RNA levels and cases with IL7R gains have been reported, the impact of IL-7Rα overexpression, rather than mutational activation, during leukemogenesis remains unclear. In this study, overexpressed IL-7Rα in tetracycline-inducible Il7r transgenic and Rosa26 IL7R knockin mice drove potential thymocyte self-renewal, and thymus hyperplasia related to increased proliferation of T-cell precursors, which subsequently infiltrated lymph nodes, spleen, and bone marrow, ultimately leading to fatal leukemia. The tumors mimicked key features of human T-ALL, including heterogeneity in immunophenotype and genetic subtype between cases, frequent hyperactivation of the PI3K/Akt pathway paralleled by downregulation of p27Kip1 and upregulation of Bcl-2, and gene expression signatures evidencing activation of JAK/STAT, PI3K/Akt/mTOR and Notch signaling. Notably, we also found that established tumors may no longer require high levels of IL-7R expression upon secondary transplantation and progressed in the absence of IL-7, but remain sensitive to inhibitors of IL-7R-mediated signaling ruxolitinib (Jak1), AZD1208 (Pim), dactolisib (PI3K/mTOR), palbociclib (Cdk4/6), and venetoclax (Bcl-2). The relevance of these findings for human disease are highlighted by the fact that samples from patients with T-ALL with high wild-type IL7R expression display a transcriptional signature resembling that of IL-7-stimulated pro-T cells and, critically, of IL7R-mutant cases of T-ALL. Overall, our study demonstrates that high expression of IL-7Rα can promote T-cell tumorigenesis, even in the absence of IL-7Rα mutational activation.
Collapse
Affiliation(s)
- Ana Silva
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Afonso R M Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Cachucho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João L Neto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sofie Demeyer
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology
- Katholieke Universiteit (KU) Leuven Center for Human Genetics, Katholieke Universiteit (VIB-KU) Leuven, Leuven, Belgium
| | - Mafalda de Matos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Thea Hogan
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Yunlei Li
- Department of Pathology Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jules Meijerink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.; and
| | - Jan Cools
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology
| | - Ana Rita Grosso
- Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Universidade NOVA de Lisboa, Caparica, Portugal
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Liang HC, Costanza M, Prutsch N, Zimmerman MW, Gurnhofer E, Montes-Mojarro IA, Abraham BJ, Prokoph N, Stoiber S, Tangermann S, Lobello C, Oppelt J, Anagnostopoulos I, Hielscher T, Pervez S, Klapper W, Zammarchi F, Silva DA, Garcia KC, Baker D, Janz M, Schleussner N, Fend F, Pospíšilová Š, Janiková A, Wallwitz J, Stoiber D, Simonitsch-Klupp I, Cerroni L, Pileri S, de Leval L, Sibon D, Fataccioli V, Gaulard P, Assaf C, Knörr F, Damm-Welk C, Woessmann W, Turner SD, Look AT, Mathas S, Kenner L, Merkel O. Super-enhancer-based identification of a BATF3/IL-2R-module reveals vulnerabilities in anaplastic large cell lymphoma. Nat Commun 2021; 12:5577. [PMID: 34552066 PMCID: PMC8458384 DOI: 10.1038/s41467-021-25379-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
Anaplastic large cell lymphoma (ALCL), an aggressive CD30-positive T-cell lymphoma, comprises systemic anaplastic lymphoma kinase (ALK)-positive, and ALK-negative, primary cutaneous and breast implant-associated ALCL. Prognosis of some ALCL subgroups is still unsatisfactory, and already in second line effective treatment options are lacking. To identify genes defining ALCL cell state and dependencies, we here characterize super-enhancer regions by genome-wide H3K27ac ChIP-seq. In addition to known ALCL key regulators, the AP-1-member BATF3 and IL-2 receptor (IL2R)-components are among the top hits. Specific and high-level IL2R expression in ALCL correlates with BATF3 expression. Confirming a regulatory link, IL-2R-expression decreases following BATF3 knockout, and BATF3 is recruited to IL2R regulatory regions. Functionally, IL-2, IL-15 and Neo-2/15, a hyper-stable IL-2/IL-15 mimic, accelerate ALCL growth and activate STAT1, STAT5 and ERK1/2. In line, strong IL-2Rα-expression in ALCL patients is linked to more aggressive clinical presentation. Finally, an IL-2Rα-targeting antibody-drug conjugate efficiently kills ALCL cells in vitro and in vivo. Our results highlight the importance of the BATF3/IL-2R-module for ALCL biology and identify IL-2Rα-targeting as a promising treatment strategy for ALCL. Anaplastic large cell lymphoma (ALCL) is an aggressive T-cell lymphoma often with poor prognosis. To identify genes defining ALCL cell state and dependencies, the authors here characterize ALCL-specific super-enhancers and describe the BATF3/IL-2R−module as a therapeutic opportunity for ALCL.
Collapse
Affiliation(s)
- Huan-Chang Liang
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK
| | - Mariantonia Costanza
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Mark W Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Elisabeth Gurnhofer
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Ivonne A Montes-Mojarro
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Institute of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nina Prokoph
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Stefan Stoiber
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory (CDL) for Applied Metabolomics, Medical University of Vienna, Vienna, Austria
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Cosimo Lobello
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Jan Oppelt
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | | | - Thomas Hielscher
- German Cancer Consortium (DKTK) German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shahid Pervez
- Department of Pathology and Laboratory Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | - Daniel-Adriano Silva
- Institute for Protein Design, University of Washington, Seattle, WA, USA.,Department of Biochemistry, University of Washington, Seattle, WA, USA.,Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, USA.,Department of Biochemistry, University of Washington, Seattle, WA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Martin Janz
- Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Nikolai Schleussner
- Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany.,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany
| | - Falko Fend
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Institute of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Šárka Pospíšilová
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic.,Department of Internal Medicine-Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Andrea Janiková
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Department of Internal Medicine-Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Jacqueline Wallwitz
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Dagmar Stoiber
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Ingrid Simonitsch-Klupp
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Lorenzo Cerroni
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Stefano Pileri
- Division of Haematopathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital (CHUV) and Lausanne University, Lausanne, Switzerland
| | - David Sibon
- Hematology Department, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, and Institut Necker-Enfants Malades, INSERM UMR1151 (Normal and pathological lymphoid differentiation), Université de Paris, Paris, France
| | - Virginie Fataccioli
- Department of Pathology, Henri Mondor University Hospital, AP-HP, INSERM U955, University Paris East, Créteil, France
| | - Philippe Gaulard
- Department of Pathology, Henri Mondor University Hospital, AP-HP, INSERM U955, University Paris East, Créteil, France
| | - Chalid Assaf
- Department of Dermatology, HELIOS Hospital Krefeld, Krefeld, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Knörr
- Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Damm-Welk
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Wilhelm Woessmann
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Suzanne D Turner
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.,Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Stephan Mathas
- European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK. .,Group Biology of Malignant Lymphomas, Max-Delbrück-Center (MDC) for Molecular Medicine, Berlin, Germany. .,Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, and Experimental and Clinical Research Center (ECRC), a joint cooperation between the MDC and Charité, Berlin, Germany. .,German Cancer Consortium (DKTK) German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Lukas Kenner
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria. .,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK. .,Christian Doppler Laboratory (CDL) for Applied Metabolomics, Medical University of Vienna, Vienna, Austria. .,Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria. .,Center for Biomarker Research in Medicine (CBMed) Core Lab 2, Medical University of Vienna, Vienna, Austria.
| | - Olaf Merkel
- Department of Pathology, Unit of Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria. .,European Research Initiative on ALK-Related Malignancies (ERIA), Suzanne Turner, Cambridge, UK.
| |
Collapse
|
23
|
Fang L, Wang W, Shi L, Chen Q, Rao X. Prognostic values and clinical relationship of TYK2 in laryngeal squamous cell cancer. Medicine (Baltimore) 2021; 100:e27062. [PMID: 34449498 PMCID: PMC10545095 DOI: 10.1097/md.0000000000027062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/20/2021] [Accepted: 08/03/2021] [Indexed: 01/10/2023] Open
Abstract
ABSTRACT Laryngeal squamous cell cancer (LSCC) is the second most common head and neck cancer with the increasing mortality. The tyrosine kinase 2 (TYK2) has previously been reported to play an important role in various cancers excepting LSCC. We used available data from the cancer genome atlas program (TCGA), gene expression omnibus, and gene expression profiling interactive analysis (GEPIA) to evaluate the role of TYK2 in LSCC.The difference of TYK2 expression level between normal and tumor samples was analyzed based on TCGA, gene expression omnibus, and GEPIA databases. The relationship between clinical features and TYK2 were analyzed using the Wilcoxon signed-rank test. We applied Cox regression and the Kaplan-Meier method to finding which clinical characteristics is associated with overall survival. Also, we used GEPIA database to validate the relationship between TYK2 and overall survival. At last, we performed gene set enrichment analysis based on TCGA data set.The expression level of TYK2 in LSCC was significantly associated with gender, lymph node status and metastasis (P-values <.05). Kaplan-Meier survival analysis, as same as GEPIA validation, demonstrated that LSCC with TYK2-low had a worse prognosis than that with TYK2-high. The univariate analysis showed that TYK2-high correlated significantly with a better overall survival (hazard ratio: 0.351, 95% confidence interval: 0.194-0.637, P < .001). The multivariate analysis revealed that TYK2 remained independently associated with overall survival (hazard ratio: 0.36, 95% confidence interval: 0.185-0.699, P = .003). Gene set enrichment analysis shows that Janus kinases-STAT signaling pathway, p53 signalling pathway and natural killer cell mediated cytotoxicity, etc are enriched in TYK2 high expression phenotype.Gene TYK2 may be a potential prognostic molecular marker for LSCC. Moreover, the Janus kinases-STAT signaling pathway and p53 signaling pathway are probably the key pathway associated with TYK2 in LC.
Collapse
Affiliation(s)
- Lucheng Fang
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Wen Wang
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Licai Shi
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Qinjuan Chen
- Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| | - Xingwang Rao
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, China
| |
Collapse
|
24
|
Borcherding DC, He K, Amin NV, Hirbe AC. TYK2 in Cancer Metastases: Genomic and Proteomic Discovery. Cancers (Basel) 2021; 13:4171. [PMID: 34439323 PMCID: PMC8393599 DOI: 10.3390/cancers13164171] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Advances in genomic analysis and proteomic tools have rapidly expanded identification of biomarkers and molecular targets important to cancer development and metastasis. On an individual basis, personalized medicine approaches allow better characterization of tumors and patient prognosis, leading to more targeted treatments by detection of specific gene mutations, overexpression, or activity. Genomic and proteomic screens by our lab and others have revealed tyrosine kinase 2 (TYK2) as an oncogene promoting progression and metastases of many types of carcinomas, sarcomas, and hematologic cancers. TYK2 is a Janus kinase (JAK) that acts as an intermediary between cytokine receptors and STAT transcription factors. TYK2 signals to stimulate proliferation and metastasis while inhibiting apoptosis of cancer cells. This review focuses on the growing evidence from genomic and proteomic screens, as well as molecular studies that link TYK2 to cancer prevalence, prognosis, and metastasis. In addition, pharmacological inhibition of TYK2 is currently used clinically for autoimmune diseases, and now provides promising treatment modalities as effective therapeutic agents against multiple types of cancer.
Collapse
Affiliation(s)
- Dana C. Borcherding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Kevin He
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Neha V. Amin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
| | - Angela C. Hirbe
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (D.C.B.); (K.H.); (N.V.A.)
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
25
|
Pocock R, Farah N, Richardson SE, Mansour MR. Current and emerging therapeutic approaches for T-cell acute lymphoblastic leukaemia. Br J Haematol 2021; 194:28-43. [PMID: 33942287 DOI: 10.1111/bjh.17310] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
T-cell ALL (T-ALL) is an aggressive malignancy of T-cell progenitors. Although survival outcomes in T-ALL have greatly improved over the past 50 years, relapsed and refractory cases remain extremely challenging to treat and those who cannot tolerate intensive treatment continue to have poor outcomes. Furthermore, T-ALL has proven a more challenging immunotherapeutic target than B-ALL. In this review we explore our expanding knowledge of the basic biology of T-ALL and how this is paving the way for repurposing established treatments and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Rachael Pocock
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Nadine Farah
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Simon E Richardson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Marc R Mansour
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
26
|
Frost ER, Ford EA, Peters AE, Reed NL, McLaughlin EA, Baker MA, Lovell-Badge R, Sutherland JM. Signal transducer and activator of transcription (STAT) 1 and STAT3 are expressed in the human ovary and have Janus kinase 1-independent functions in the COV434 human granulosa cell line. Reprod Fertil Dev 2021; 32:1027-1039. [PMID: 32758351 DOI: 10.1071/rd20098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/29/2020] [Indexed: 01/03/2023] Open
Abstract
Ovarian granulosa cells are fundamental for oocyte maintenance and maturation. Recent studies have demonstrated the importance of members of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway in the granulosa cell population of mouse and horse ovaries, with perturbation of JAK1 signalling in the mouse shown to impair oocyte maintenance and accelerate primordial follicle activation. The presence and role of the JAK/STAT pathway in human granulosa cells has yet to be elucidated. In this study, expression of JAK1, STAT1 and STAT3 was detected in oocytes and granulosa cells of human ovarian sections from fetal (40 weeks gestation) and premenopausal ovaries (34-41 years of age; n=3). To determine the effects of JAK1 signalling in granulosa cells, the human granulosa-like cell line COV434 was used, with JAK1 inhibition using ruxolitinib. Chemical inhibition of JAK1 in COV434 cells with 100nM ruxolitinib for 72h resulted in significant increases in STAT3 mRNA (P=0.034) and p-Y701-STAT1 protein (P=0.0117), demonstrating a role for JAK1 in modulating STAT in granulosa cells. This study implicates a conserved role for JAK/STAT signalling in human ovary development, warranting further investigation of this pathway in human granulosa cell function.
Collapse
Affiliation(s)
- E R Frost
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; and Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; and Corresponding author.
| | - E A Ford
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - A E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - N L Reed
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - E A McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and School of Science, Western Sydney University, Penrith, NSW 2751, Australia; and School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand
| | - M A Baker
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - R Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - J M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
27
|
Untwining Anti-Tumor and Immunosuppressive Effects of JAK Inhibitors-A Strategy for Hematological Malignancies? Cancers (Basel) 2021; 13:cancers13112611. [PMID: 34073410 PMCID: PMC8197909 DOI: 10.3390/cancers13112611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is aberrantly activated in many malignancies. Inhibition of this pathway via JAK inhibitors (JAKinibs) is therefore an attractive therapeutic strategy underlined by Ruxolitinib (JAK1/2 inhibitor) being approved for the treatment of myeloproliferative neoplasms. As a consequence of the crucial role of the JAK-STAT pathway in the regulation of immune responses, inhibition of JAKs suppresses the immune system. This review article provides a thorough overview of the current knowledge on JAKinibs’ effects on immune cells in the context of hematological malignancies. We also discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of the malignancy. Abstract The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway propagates signals from a variety of cytokines, contributing to cellular responses in health and disease. Gain of function mutations in JAKs or STATs are associated with malignancies, with JAK2V617F being the main driver mutation in myeloproliferative neoplasms (MPN). Therefore, inhibition of this pathway is an attractive therapeutic strategy for different types of cancer. Numerous JAK inhibitors (JAKinibs) have entered clinical trials, including the JAK1/2 inhibitor Ruxolitinib approved for the treatment of MPN. Importantly, loss of function mutations in JAK-STAT members are a cause of immune suppression or deficiencies. MPN patients undergoing Ruxolitinib treatment are more susceptible to infections and secondary malignancies. This highlights the suppressive effects of JAKinibs on immune responses, which renders them successful in the treatment of autoimmune diseases but potentially detrimental for cancer patients. Here, we review the current knowledge on the effects of JAKinibs on immune cells in the context of hematological malignancies. Furthermore, we discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of malignancies. In summary, this review underlines the necessity of a robust immune profiling to provide the best benefit for JAKinib-treated patients.
Collapse
|
28
|
Walker KL, Rinella SP, Hess NJ, Turicek DP, Kabakov SA, Zhu F, Bouchlaka MN, Olson SL, Cho MM, Quamine AE, Feils AS, Gavcovich TB, Rui L, Capitini CM. CXCR4 allows T cell acute lymphoblastic leukemia to escape from JAK1/2 and BCL2 inhibition through CNS infiltration. Leuk Lymphoma 2021; 62:1167-1177. [PMID: 33843403 DOI: 10.1080/10428194.2021.1910684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Targeting the JAK/STAT and BCL2 pathways in patients with relapsed/refractory T cell acute lymphoblastic leukemia (T-ALL) may provide an alternative approach to achieve clinical remissions. Ruxolitinib and venetoclax show a dose-dependent effect on T-ALL individually, but combination treatment reduces survival and proliferation of T-ALL in vitro. Using a xenograft model, the combination treatment fails to improve survival, with death from hind limb paralysis. Despite on-target inhibition by the drugs, histopathology demonstrates increased leukemic infiltration into the central nervous system (CNS) as compared to liver or bone marrow. Liquid chromatography-tandem mass spectroscopy shows that ruxolitinib and venetoclax insufficiently cross into the CNS. The addition of the CXCR4 inhibitor plerixafor with ruxolitinib and venetoclax reduces clinical scores and enhances survival. While combination therapy with ruxolitinib and venetoclax shows promise for treating T-ALL, additional inhibition of the CXCR4-CXCL12 axis may be needed to maximize the possibility of complete remission.
Collapse
Affiliation(s)
- Kirsti L Walker
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean P Rinella
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nicholas J Hess
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David P Turicek
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sabrina A Kabakov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Fen Zhu
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Myriam N Bouchlaka
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sydney L Olson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Monica M Cho
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Aicha E Quamine
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Arika S Feils
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tara B Gavcovich
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lixin Rui
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
29
|
Jia X, Huang C, Hu Y, Wu Q, Liu F, Nie W, Chen H, Li X, Dong Z, Liu K. Cirsiliol targets tyrosine kinase 2 to inhibit esophageal squamous cell carcinoma growth in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:105. [PMID: 33731185 PMCID: PMC7972218 DOI: 10.1186/s13046-021-01903-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/08/2021] [Indexed: 12/19/2022]
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is an aggressive and lethal cancer with a low 5 year survival rate. Identification of new therapeutic targets and its inhibitors remain essential for ESCC prevention and treatment. Methods TYK2 protein levels were checked by immunohistochemistry. The function of TYK2 in cell proliferation was investigated by MTT [(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] and anchorage-independent cell growth. Computer docking, pull-down assay, surface plasmon resonance, and kinase assay were used to confirm the binding and inhibition of TYK2 by cirsiliol. Cell proliferation, western blot and patient-derived xenograft tumor model were used to determine the inhibitory effects and mechanism of cirsiliol in ESCC. Results TYK2 was overexpressed and served as an oncogene in ESCC. Cirsiliol could bind with TYK2 and inhibit its activity, thereby decreasing dimer formation and nucleus localization of signal transducer and activator of transcription 3 (STAT3). Cirsiliol could inhibit ESCC growth in vitro and in vivo. Conclusions TYK2 is a potential target in ESCC, and cirsiliol could inhibit ESCC by suppression of TYK2. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01903-z.
Collapse
Affiliation(s)
- Xuechao Jia
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Chuntian Huang
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Yamei Hu
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Qiong Wu
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Fangfang Liu
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Xiang Li
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China
| | - Zigang Dong
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China.
| | - Kangdong Liu
- Department of Pathophysiology, The School of Basic Medical Sciences, AMS, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450008, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
| |
Collapse
|
30
|
Janus Kinases in Leukemia. Cancers (Basel) 2021; 13:cancers13040800. [PMID: 33672930 PMCID: PMC7918039 DOI: 10.3390/cancers13040800] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 01/12/2023] Open
Abstract
Janus kinases (JAKs) transduce signals from dozens of extracellular cytokines and function as critical regulators of cell growth, differentiation, gene expression, and immune responses. Deregulation of JAK/STAT signaling is a central component in several human diseases including various types of leukemia and other malignancies and autoimmune diseases. Different types of leukemia harbor genomic aberrations in all four JAKs (JAK1, JAK2, JAK3, and TYK2), most of which are activating somatic mutations and less frequently translocations resulting in constitutively active JAK fusion proteins. JAKs have become important therapeutic targets and currently, six JAK inhibitors have been approved by the FDA for the treatment of both autoimmune diseases and hematological malignancies. However, the efficacy of the current drugs is not optimal and the full potential of JAK modulators in leukemia is yet to be harnessed. This review discusses the deregulation of JAK-STAT signaling that underlie the pathogenesis of leukemia, i.e., mutations and other mechanisms causing hyperactive cytokine signaling, as well as JAK inhibitors used in clinic and under clinical development.
Collapse
|
31
|
Takao S, Forbes L, Uni M, Cheng S, Pineda JMB, Tarumoto Y, Cifani P, Minuesa G, Chen C, Kharas MG, Bradley RK, Vakoc CR, Koche RP, Kentsis A. Convergent organization of aberrant MYB complex controls oncogenic gene expression in acute myeloid leukemia. eLife 2021; 10:e65905. [PMID: 33527899 PMCID: PMC7886351 DOI: 10.7554/elife.65905] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulated gene expression contributes to most prevalent features in human cancers. Here, we show that most subtypes of acute myeloid leukemia (AML) depend on the aberrant assembly of MYB transcriptional co-activator complex. By rapid and selective peptidomimetic interference with the binding of CBP/P300 to MYB, but not CREB or MLL1, we find that the leukemic functions of MYB are mediated by CBP/P300 co-activation of a distinct set of transcription factor complexes. These MYB complexes assemble aberrantly with LYL1, E2A, C/EBP family members, LMO2, and SATB1. They are organized convergently in genetically diverse subtypes of AML and are at least in part associated with inappropriate transcription factor co-expression. Peptidomimetic remodeling of oncogenic MYB complexes is accompanied by specific proteolysis and dynamic redistribution of CBP/P300 with alternative transcription factors such as RUNX1 to induce myeloid differentiation and apoptosis. Thus, aberrant assembly and sequestration of MYB:CBP/P300 complexes provide a unifying mechanism of oncogenic gene expression in AML. This work establishes a compelling strategy for their pharmacologic reprogramming and therapeutic targeting for diverse leukemias and possibly other human cancers caused by dysregulated gene control.
Collapse
Affiliation(s)
- Sumiko Takao
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Lauren Forbes
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Departments of Pharmacology and Physiology & Biophysics, Weill Cornell Graduate School of Medical Sciences, Cornell UniversityNew YorkUnited States
| | - Masahiro Uni
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Shuyuan Cheng
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Departments of Pharmacology and Physiology & Biophysics, Weill Cornell Graduate School of Medical Sciences, Cornell UniversityNew YorkUnited States
| | - Jose Mario Bello Pineda
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Medical Scientist Training Program, University of WashingtonSeattleUnited States
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Yusuke Tarumoto
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Institute for Frontier Life and Medical Sciences, Kyoto UniversityKyotoJapan
| | - Paolo Cifani
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Gerard Minuesa
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Celine Chen
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Michael G Kharas
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Departments of Pharmacology and Physiology & Biophysics, Weill Cornell Graduate School of Medical Sciences, Cornell UniversityNew YorkUnited States
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | | | - Richard P Koche
- Center for Epigenetics Research, Sloan Kettering InstituteNew YorkUnited States
| | - Alex Kentsis
- Molecular Pharmacology Program, Sloan Kettering InstituteNew YorkUnited States
- Tow Center for Developmental Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Departments of Pharmacology and Physiology & Biophysics, Weill Cornell Graduate School of Medical Sciences, Cornell UniversityNew YorkUnited States
| |
Collapse
|
32
|
Klco JM, Mullighan CG. Advances in germline predisposition to acute leukaemias and myeloid neoplasms. Nat Rev Cancer 2021; 21:122-137. [PMID: 33328584 PMCID: PMC8404376 DOI: 10.1038/s41568-020-00315-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Although much work has focused on the elucidation of somatic alterations that drive the development of acute leukaemias and other haematopoietic diseases, it has become increasingly recognized that germline mutations are common in many of these neoplasms. In this Review, we highlight the different genetic pathways impacted by germline mutations that can ultimately lead to the development of familial and sporadic haematological malignancies, including acute lymphoblastic leukaemia, acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS). Many of the genes disrupted by somatic mutations in these diseases (for example, TP53, RUNX1, IKZF1 and ETV6) are the same as those that harbour germline mutations in children and adolescents who develop these malignancies. Moreover, the presumption that familial leukaemias only present in childhood is no longer true, in large part due to the numerous studies demonstrating germline DDX41 mutations in adults with MDS and AML. Lastly, we highlight how different cooperating events can influence the ultimate phenotype in these different familial leukaemia syndromes.
Collapse
Affiliation(s)
- Jeffery M Klco
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Charles G Mullighan
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
33
|
Favoino E, Prete M, Catacchio G, Ruscitti P, Navarini L, Giacomelli R, Perosa F. Working and safety profiles of JAK/STAT signaling inhibitors. Are these small molecules also smart? Autoimmun Rev 2021; 20:102750. [PMID: 33482338 DOI: 10.1016/j.autrev.2021.102750] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022]
Abstract
The Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway is an important intracellular route through which many different extracellular soluble molecules, by reaching membrane receptors, can signal the nucleus. The spectrum of soluble molecules that use the JAK/STAT pathway through their corresponding receptors is quite large (almost 50 different molecules), and includes some cytokines involved in the pathogenesis of many immune-mediated diseases. Such diseases, when left untreated, present an evident hyperactivation of JAK/STAT signaling. Therefore, given the pathogenetic role of JAK/STAT, drugs known as JAK inhibitors (JAKi), that target one or more JAKs, have been developed to counteract JAK/STAT signal hyperactivation. As some hematological malignancies present an intrinsic JAK/STAT hyperactivation due to a JAK mutation, some JAKi have also been successfully used in this context. Regulatory agencies for drug administration in different countries have already approved a few JAKi in the setting of either immune-mediated diseases or hematological malignancies. Aim of this review is to describe the physiology of intracellular JAK/STAT pathway signaling and the pathological conditions associated to its dysregulation. Then, the rationale for targeting JAK in rheumatic autoimmune diseases is discussed, along with clinical data from registration studies showing the efficacy of these drugs. Finally, the excellent safety profile of JAKi is discussed in the context of the apparent poor specificity of JAK/STAT pathway signal.
Collapse
Affiliation(s)
- Elvira Favoino
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Marcella Prete
- Internal Medicine, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Giacomo Catacchio
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Luca Navarini
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome 'Campus Biomedico', Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome 'Campus Biomedico', Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy.
| |
Collapse
|
34
|
Acute lymphoid leukemia etiopathogenesis. Mol Biol Rep 2021; 48:817-822. [PMID: 33438082 DOI: 10.1007/s11033-020-06073-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
Acute lymphoid leukemia (ALL) is a type of hematological neoplasm that affects the precursor cells of strains B, T and NK, with a higher incidence in the pediatric range. The pathophysiology of ALL is characterized by chromosomal abnormalities and genetic alterations involved in the differentiation and proliferation of lymphoid precursor cells. Despite the lack of information in the literature, it is believed that leukemogenesis originates from a complex interaction between environmental and genetic factors, which combined lead to cellular modifications. Environmental factors have been evaluated as possible predisposing factors in the development of ALL but there are still conflicting results in the world literature. In this context, the aim of the present review is to discuss the major exogenous factors regarding ALL.
Collapse
|
35
|
TYK2 Variants in B-Acute Lymphoblastic Leukaemia. Genes (Basel) 2020; 11:genes11121434. [PMID: 33260630 PMCID: PMC7761059 DOI: 10.3390/genes11121434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/31/2022] Open
Abstract
B-cell precursor acute lymphoblastic leukaemia (B-ALL) is a malignancy of lymphoid progenitor cells with altered genes including the Janus kinase (JAK) gene family. Among them, tyrosine kinase 2 (TYK2) is involved in signal transduction of cytokines such as interferon (IFN) α/β through IFN−α/β receptor alpha chain (IFNAR1). To search for disease-associated TYK2 variants, bone marrow samples from 62 B-ALL patients at diagnosis were analysed by next-generation sequencing. TYK2 variants were found in 16 patients (25.8%): one patient had a novel mutation at the four-point-one, ezrin, radixin, moesin (FERM) domain (S431G) and two patients had the rare variants rs150601734 or rs55882956 (R425H or R832W). To functionally characterise them, they were generated by direct mutagenesis, cloned in expression vectors, and transfected in TYK2-deficient cells. Under high-IFNα doses, the three variants were competent to phosphorylate STAT1/2. While R425H and R832W induced STAT1/2-target genes measured by qPCR, S431G behaved as the kinase-dead form of the protein. None of these variants phosphorylated STAT3 in in vitro kinase assays. Molecular dynamics simulation showed that TYK2/IFNAR1 interaction is not affected by these variants. Finally, qPCR analysis revealed diminished expression of TYK2 in B-ALL patients at diagnosis compared to that in healthy donors, further stressing the tumour immune surveillance role of TYK2.
Collapse
|
36
|
Meltendorf S, Fu H, Pierau M, Lindquist JA, Finzel S, Mertens PR, Gieseler-Halbach S, Ambach A, Thomas U, Lingel H, Voll RE, Brunner-Weinzierl MC. Cell Survival Failure in Effector T Cells From Patients With Systemic Lupus Erythematosus Following Insufficient Up-Regulation of Cold-Shock Y-Box Binding Protein 1. Arthritis Rheumatol 2020; 72:1721-1733. [PMID: 32475063 DOI: 10.1002/art.41382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/23/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The importance of cold-shock Y-box binding protein 1 (YB-1) for cell homeostasis is well-documented based on prior observations of its association with certain cancer entities. This study was undertaken to explore the role of YB-1 in T cell homeostasis and survival and the potential contribution of YB-1 to the pathogenesis of systemic lupus erythematosus (SLE). METHODS In the peripheral blood from 25 SLE patients and 25 healthy donors, the expression of YB-1 and frequency of T cell apoptosis was analyzed by quantitative polymerase chain reaction (qPCR) and fluorescence-activated cell sorting of CD4+ T cells ex vivo and also analyzed in T cells in vitro after 6 days of stimulation with anti-CD3-coupled or anti-CD3/anti-CD28-coupled microspheres. YB-1 was overexpressed using lentiviral transduction with wild-type green fluorescent protein (wtGFP) YB-1, and knockdown of YB-1 was achieved using specific short hairpin RNA (shRNA) (3-fold reduction; P < 0.0001). RESULTS YB-1 expression was significantly lower in apoptosis-prone T cells and in activated T cells from SLE patients compared to YB-1 expression in nonapoptotic T cells and activated T cells from healthy donors (P = 0.001). Knockdown of YB-1 in T cells consequently led to expression of proapoptotic molecules and caspase 3 activation (1.6-fold), and subsequently, to apoptosis. Furthermore, YB-1 promoted survival pathways involving enhanced protein expression of the kinase Akt (2-fold) and Bcl-2 (3-fold), even when Fas/CD95 was triggered. YB-1-mediated T cell survival was reversed by Akt and phosphatidylinositol 3-kinase (PI3K) inactivation. In SLE patients, rescue of YB-1 expression strongly promoted survival of T cells and even prevented cell death in T cells that were extremely apoptosis-prone. CONCLUSION Our data show that failure of YB-1 up-regulation in T cells from SLE patients led to enhanced apoptosis. These findings imply that YB-1 plays a crucial role in the disturbed homeostasis of activated T cells leading to hematopoietic alterations in SLE. These insights may help facilitate the development of new treatment strategies for SLE.
Collapse
Affiliation(s)
- Stefan Meltendorf
- Department of Experimental Pediatrics, Otto von uericke University, Magdeburg, Germany
| | - Hang Fu
- Department of Experimental Pediatrics, Otto von uericke University, Magdeburg, Germany
| | - Mandy Pierau
- Department of Experimental Pediatrics, Otto von uericke University, Magdeburg, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology, Hypertension, Diabetes, and Endocrinology, Otto von Guericke University, Magdeburg, Germany
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Albert Ludwig University of Freiburg, Freiburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology, Hypertension, Diabetes, and Endocrinology, Otto von Guericke University, Magdeburg, Germany
| | | | - Andreas Ambach
- Department of Dermatology, Otto von Guericke University, Magdeburg, Germany
| | - Ulrich Thomas
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, Otto von uericke University, Magdeburg, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Albert Ludwig University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
37
|
Prudner BC, Ball T, Rathore R, Hirbe AC. Diagnosis and management of malignant peripheral nerve sheath tumors: Current practice and future perspectives. Neurooncol Adv 2020; 2:i40-i49. [PMID: 32642731 PMCID: PMC7317062 DOI: 10.1093/noajnl/vdz047] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
One of the most common malignancies affecting adults with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome is the malignant peripheral nerve sheath tumor (MPNST), a highly aggressive sarcoma that typically develops from benign plexiform neurofibromas. Approximately 8-13% of individuals with NF1 will develop MPNST during young adulthood. There are few therapeutic options, and the vast majority of people with these cancers will die within 5 years of diagnosis. Despite efforts to understand the pathogenesis of these aggressive tumors, the overall prognosis remains dismal. This manuscript will review the current understanding of the cellular and molecular progression of MPNST, diagnostic workup of patients with these tumors, current treatment paradigms, and investigational treatment options. Additionally, we highlight novel areas of preclinical research, which may lead to future clinical trials. In summary, MPNST remains a diagnostic and therapeutic challenge, and future work is needed to develop novel and rational combinational therapy for these tumors.
Collapse
Affiliation(s)
- Bethany C Prudner
- Division of Medical Oncology, Department of Medicine, Washington University, St. Louis
| | - Tyler Ball
- Division of Medical Oncology, Department of Medicine, Washington University, St. Louis
| | - Richa Rathore
- Division of Medical Oncology, Department of Medicine, Washington University, St. Louis
| | - Angela C Hirbe
- Division of Medical Oncology, Department of Medicine, Washington University, St. Louis
- Neurofibromatosis Center, Washington University, St. Louis MO
- Siteman Cancer Center, Washington University, St. Louis
| |
Collapse
|
38
|
Perera Y, Melão A, Ramón AC, Vázquez D, Ribeiro D, Perea SE, Barata JT. Clinical-Grade Peptide-Based Inhibition of CK2 Blocks Viability and Proliferation of T-ALL Cells and Counteracts IL-7 Stimulation and Stromal Support. Cancers (Basel) 2020; 12:cancers12061377. [PMID: 32471246 PMCID: PMC7352628 DOI: 10.3390/cancers12061377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023] Open
Abstract
Despite remarkable advances in the treatment of T-cell acute lymphoblastic leukemia (T-ALL), relapsed cases are still a major challenge. Moreover, even successful cases often face long-term treatment-associated toxicities. Targeted therapeutics may overcome these limitations. We have previously demonstrated that casein kinase 2 (CK2)-mediated phosphatase and tensin homologue (PTEN) posttranslational inactivation, and consequent phosphatidylinositol 3-kinase (PI3K)/Akt signaling hyperactivation, leads to increased T-ALL cell survival and proliferation. We also revealed the existence of a crosstalk between CK2 activity and the signaling mediated by interleukin 7 (IL-7), a critical leukemia-supportive cytokine. Here, we evaluated the impact of CIGB-300, a the clinical-grade peptide-based CK2 inhibitor CIGB-300 on T-ALL biology. We demonstrate that CIGB-300 decreases the viability and proliferation of T-ALL cell lines and diagnostic patient samples. Moreover, CIGB-300 overcomes IL-7-mediated T-ALL cell growth and viability, while preventing the positive effects of OP9-delta-like 1 (DL1) stromal support on leukemia cells. Signaling and pull-down experiments indicate that the CK2 substrate nucleophosmin 1 (B23/NPM1) and CK2 itself are the molecular targets for CIGB-300 in T-ALL cells. However, B23/NPM1 silencing only partially recapitulates the anti-leukemia effects of the peptide, suggesting that CIGB-300-mediated direct binding to CK2, and consequent CK2 inactivation, is the mechanism by which CIGB-300 downregulates PTEN S380 phosphorylation and inhibits PI3K/Akt signaling pathway. In the context of IL-7 stimulation, CIGB-300 blocks janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in T-ALL cells. Altogether, our results strengthen the case for anti-CK2 therapeutic intervention in T-ALL, demonstrating that CIGB-300 (given its ability to circumvent the effects of pro-leukemic microenvironmental cues) may be a valid tool for clinical intervention in this aggressive malignancy.
Collapse
Affiliation(s)
- Yasser Perera
- Laboratory of Molecular Oncology, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (Y.P.); (A.C.R.); (S.E.P.)
| | - Alice Melão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (A.M.); (D.R.)
| | - Ailyn C. Ramón
- Laboratory of Molecular Oncology, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (Y.P.); (A.C.R.); (S.E.P.)
| | - Dania Vázquez
- Pharmacogenomics Department, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba;
| | - Daniel Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (A.M.); (D.R.)
| | - Silvio E. Perea
- Laboratory of Molecular Oncology, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba; (Y.P.); (A.C.R.); (S.E.P.)
| | - João T. Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal; (A.M.); (D.R.)
- Correspondence:
| |
Collapse
|
39
|
The effect of co-occurring lesions on leukaemogenesis and drug response in T-ALL and ETP-ALL. Br J Cancer 2019; 122:455-464. [PMID: 31792348 PMCID: PMC7028932 DOI: 10.1038/s41416-019-0647-7] [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] [Received: 06/25/2019] [Revised: 10/14/2019] [Accepted: 10/30/2019] [Indexed: 01/27/2023] Open
Abstract
Despite advances in the management of acute lymphoblastic leukaemia (ALL), current regimens fail to significantly transform outcomes for patients with high-risk subtypes. Advances in genomic analyses have identified novel lesions including mutations in genes that encode chromatin modifiers and those that influence cytokine and kinase signalling, rendering many of these alterations potentially targetable by tyrosine kinase and epigenetic inhibitors currently in clinical use. Although specific genomic lesions, gene expression patterns, and immunophenotypic profiles have been associated with specific clinical outcomes in some cancers, the application of precision medicine approaches based on these data has been slow. This approach is complicated by the reality that patients often harbour multiple mutations, and in many cases, the precise functional significance and interaction of these mutations in driving leukaemia and drug responsiveness/resistance remains unknown. Given that signalling pathways driving leukaemic pathogenesis could plausibly result from the co-existence of specific lesions and the resultant perturbation of protein interactions, the use of combined therapeutics that target multiple aberrant pathways, according to an individual’s mutational profile, might improve outcomes and lower a patient’s risk of relapse. Here we outline the genomic alterations that occur in T cell ALL (T-ALL) and early T cell precursor (ETP)-ALL and review studies highlighting the possible effects of co-occurring lesions on leukaemogenesis and drug response.
Collapse
|
40
|
Orlova A, Wagner C, de Araujo ED, Bajusz D, Neubauer HA, Herling M, Gunning PT, Keserű GM, Moriggl R. Direct Targeting Options for STAT3 and STAT5 in Cancer. Cancers (Basel) 2019; 11:E1930. [PMID: 31817042 PMCID: PMC6966570 DOI: 10.3390/cancers11121930] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Signal transducer and activator of transcription (STAT)3 and STAT5 are important transcription factors that are able to mediate or even drive cancer progression through hyperactivation or gain-of-function mutations. Mutated STAT3 is mainly associated with large granular lymphocytic T-cell leukemia, whereas mutated STAT5B is associated with T-cell prolymphocytic leukemia, T-cell acute lymphoblastic leukemia and γδ T-cell-derived lymphomas. Hyperactive STAT3 and STAT5 are also implicated in various hematopoietic and solid malignancies, such as chronic and acute myeloid leukemia, melanoma or prostate cancer. Classical understanding of STAT functions is linked to their phosphorylated parallel dimer conformation, in which they induce gene transcription. However, the functions of STAT proteins are not limited to their phosphorylated dimerization form. In this review, we discuss the functions and the roles of unphosphorylated STAT3/5 in the context of chromatin remodeling, as well as the impact of STAT5 oligomerization on differential gene expression in hematopoietic neoplasms. The central involvement of STAT3/5 in cancer has made these molecules attractive targets for small-molecule drug development, but currently there are no direct STAT3/5 inhibitors of clinical grade available. We summarize the development of inhibitors against the SH2 domains of STAT3/5 and discuss their applicability as cancer therapeutics.
Collapse
Affiliation(s)
- Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Christina Wagner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Elvin D. de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center for Molecular Medicine Cologne (CMMC), Cologne University, 50937 Cologne, Germany;
| | - Patrick T. Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - György M. Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| |
Collapse
|
41
|
|
42
|
Wöss K, Simonović N, Strobl B, Macho-Maschler S, Müller M. TYK2: An Upstream Kinase of STATs in Cancer. Cancers (Basel) 2019; 11:E1728. [PMID: 31694222 PMCID: PMC6896190 DOI: 10.3390/cancers11111728] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023] Open
Abstract
In this review we concentrate on the recent findings describing the oncogenic potential of the protein tyrosine kinase 2 (TYK2). The overview on the current understanding of TYK2 functions in cytokine responses and carcinogenesis focusses on the activation of the signal transducers and activators of transcription (STAT) 3 and 5. Insight gained from loss-of-function (LOF) gene-modified mice and human patients homozygous for Tyk2/TYK2-mutated alleles established the central role in immunological and inflammatory responses. For the description of physiological TYK2 structure/function relationships in cytokine signaling and of overarching molecular and pathologic properties in carcinogenesis, we mainly refer to the most recent reviews. Dysregulated TYK2 activation, aberrant TYK2 protein levels, and gain-of-function (GOF) TYK2 mutations are found in various cancers. We discuss the molecular consequences thereof and briefly describe the molecular means to counteract TYK2 activity under (patho-)physiological conditions by cellular effectors and by pharmacological intervention. For the role of TYK2 in tumor immune-surveillance we refer to the recent Special Issue of Cancers "JAK-STAT Signaling Pathway in Cancer".
Collapse
Affiliation(s)
| | | | | | | | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria; (K.W.); (N.S.); (B.S.); (S.M.-M.)
| |
Collapse
|
43
|
Catuogno S, Di Martino MT, Nuzzo S, Esposito CL, Tassone P, de Franciscis V. An Anti-BCMA RNA Aptamer for miRNA Intracellular Delivery. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:981-990. [PMID: 31778956 PMCID: PMC6889555 DOI: 10.1016/j.omtn.2019.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022]
Abstract
B cell maturation antigen is highly expressed on malignant plasma cells in human multiple myeloma and has recently emerged as a very promising target for therapeutic interventions. Nucleic-acid-based aptamers are small oligonucleotides with high selective targeting properties and functional advantages over monoclonal antibodies, as both diagnostic and therapeutic tools. Here, we describe the generation of the first-ever-described nuclease resistant RNA aptamer selectively binding to B cell maturation antigen. We adopted a modified cell-based systematic evolution of ligands by exponential enrichment approach allowing the enrichment for internalizing aptamers. The selected 2′Fluoro-Pyrimidine modified aptamer, named apt69.T, effectively and selectively bound B cell maturation antigen-expressing myeloma cells with rapid and efficient internalization. Interestingly, apt69.T inhibited APRIL-dependent nuclear factor κB (NF-κB) pathway in vitro. Moreover, the aptamer was conjugated to microRNA-137 (miR-137) and anti-miR-222, demonstrating high potential against tumor cells. In conclusion, apt69.T is a novel tool suitable for direct targeting and delivery of therapeutics to B cell maturation antigen-expressing myeloma cells.
Collapse
Affiliation(s)
- Silvia Catuogno
- IEOS - Istituto per l'endocrinologia e l'oncologia "Gaetano Salvatore," CNR, Naples, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | | | - Carla Lucia Esposito
- IEOS - Istituto per l'endocrinologia e l'oncologia "Gaetano Salvatore," CNR, Naples, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy.
| | - Vittorio de Franciscis
- IEOS - Istituto per l'endocrinologia e l'oncologia "Gaetano Salvatore," CNR, Naples, Italy.
| |
Collapse
|
44
|
Paganelli F, Lonetti A, Anselmi L, Martelli AM, Evangelisti C, Chiarini F. New advances in targeting aberrant signaling pathways in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2019; 74:100649. [PMID: 31523031 DOI: 10.1016/j.jbior.2019.100649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disorder characterized by malignant transformation of immature progenitors primed towards T-cell development. Over the past 15 years, advances in the molecular characterization of T-ALL have uncovered oncogenic key drivers and crucial signaling pathways of this disease, opening new chances for the development of novel therapeutic strategies. Currently, T-ALL patients are still treated with aggressive therapies, consisting of high dose multiagent chemotherapy. To minimize and overcome the unfavorable effects of these regimens, it is critical to identify innovative targets and test selective inhibitors of such targets. Major efforts are being made to develop small molecules against deregulated signaling pathways, which sustain T-ALL cell growth, survival, metabolism, and drug-resistance. This review will focus on recent improvements in the understanding of the signaling pathways involved in the pathogenesis of T-ALL and on the challenging opportunities for T-ALL targeted therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Annalisa Lonetti
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Laura Anselmi
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Chiarini
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
45
|
Qin W, Godec A, Zhang X, Zhu C, Shao J, Tao Y, Bu X, Hirbe AC. TYK2 promotes malignant peripheral nerve sheath tumor progression through inhibition of cell death. Cancer Med 2019; 8:5232-5241. [PMID: 31278855 PMCID: PMC6718590 DOI: 10.1002/cam4.2386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/05/2019] [Accepted: 06/18/2019] [Indexed: 01/01/2023] Open
Abstract
Background Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that arise most commonly in the setting of the Neurofibromatosis Type 1 (NF1) cancer predisposition syndrome. Despite aggressive multimodality therapy, outcomes are dismal and most patients die within 5 years of diagnosis. Prior genomic studies in our laboratory identified tyrosine kinase 2 (TYK2) as a frequently mutated gene in MPNST. Herein, we explored the function of TYK2 in MPNST pathogenesis. Methods Immunohistochemistry was utilized to examine expression of TYK2 in MPNSTs and other sarcomas. To establish a role for TYK2 in MPNST pathogenesis, murine and human TYK2 knockdown and knockout cells were established using shRNA and CRISPR/Cas9 systems, respectively. Results We have demonstrated that TYK2 was highly expressed in the majority of human MPNSTs examined. Additionally, we demonstrated that knockdown of Tyk2/TYK2 in murine and human MPNST cells significantly increased cell death in vitro. These effects were accompanied by a decrease in the levels of activated Stats and Bcl‐2 as well as an increase in the levels of Cleaved Caspase‐3. In addition, Tyk2‐KD cells demonstrated impaired growth in subcutaneous and metastasis models in vivo. Conclusion Taken together, these data illustrate the importance of TYK2 in MPNST pathogenesis and suggest that the TYK2 pathway may be a potential therapeutic target for these deadly cancers.
Collapse
Affiliation(s)
- Wenjing Qin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.,School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Abigail Godec
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaochun Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Cuige Zhu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jieya Shao
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri
| | - Yu Tao
- Cancer Center Biostatistics Shared Resource, Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Angela C Hirbe
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
46
|
Blazanin N, Cheng T, Carbajal S, DiGiovanni J. Activation of a protumorigenic IFNγ/STAT1/IRF‐1 signaling pathway in keratinocytes following exposure to solar ultraviolet light. Mol Carcinog 2019; 58:1656-1669. [DOI: 10.1002/mc.23073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Nicholas Blazanin
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at Austin Austin Texas
| | - Tianyi Cheng
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at Austin Austin Texas
| | - Steve Carbajal
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at Austin Austin Texas
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of PharmacyThe University of Texas at Austin Austin Texas
- Department of Pediatrics, Dell Medical SchoolThe University of Texas at Austin Austin Texas
- Center for Molecular Carcinogenesis and ToxicologyThe University of Texas at Austin Austin Texas
| |
Collapse
|
47
|
Hammarén HM, Virtanen AT, Raivola J, Silvennoinen O. The regulation of JAKs in cytokine signaling and its breakdown in disease. Cytokine 2019; 118:48-63. [DOI: 10.1016/j.cyto.2018.03.041] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 01/12/2023]
|
48
|
Sharma ND, Nickl CK, Kang H, Ornatowski W, Brown R, Ness SA, Loh ML, Mullighan CG, Winter SS, Hunger SP, Cannon JL, Matlawska‐Wasowska K. Epigenetic silencing of SOCS5 potentiates JAK-STAT signaling and progression of T-cell acute lymphoblastic leukemia. Cancer Sci 2019; 110:1931-1946. [PMID: 30974024 PMCID: PMC6549933 DOI: 10.1111/cas.14021] [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: 09/27/2018] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
Activating mutations in cytokine receptors and transcriptional regulators govern aberrant signal transduction in T-cell lineage acute lymphoblastic leukemia (T-ALL). However, the roles played by suppressors of cytokine signaling remain incompletely understood. We examined the regulatory roles of suppressor of cytokine signaling 5 (SOCS5) in T-ALL cellular signaling networks and leukemia progression. We found that SOCS5 was differentially expressed in primary T-ALL and its expression levels were lowered in HOXA-deregulated leukemia harboring KMT2A gene rearrangements. Here, we report that SOCS5 expression is epigenetically regulated by DNA methyltransferase-3A-mediated DNA methylation and methyl CpG binding protein-2-mediated histone deacetylation. We show that SOCS5 negatively regulates T-ALL cell growth and cell cycle progression but has no effect on apoptotic cell death. Mechanistically, SOCS5 silencing induces activation of JAK-STAT signaling, and negatively regulates interleukin-7 and interleukin-4 receptors. Using a human T-ALL murine xenograft model, we show that genetic inactivation of SOCS5 accelerates leukemia engraftment and progression, and leukemia burden. We postulate that SOCS5 is epigenetically deregulated in T-ALL and serves as an important regulator of T-ALL cell proliferation and leukemic progression. Our results link aberrant downregulation of SOCS5 expression to the enhanced activation of the JAK-STAT and cytokine receptor-signaling cascade in T-ALL.
Collapse
Affiliation(s)
- Nitesh D. Sharma
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Christian K. Nickl
- Department of PediatricsUniversity of New Mexico Health Sciences CenterAlbuquerqueNM
| | - Huining Kang
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Wojciech Ornatowski
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Roger Brown
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Scott A. Ness
- Department of Internal MedicineUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
| | - Mignon L. Loh
- Department of PediatricsBenioff Children's HospitalUniversity of California at San FranciscoSan FranciscoCA
| | | | - Stuart S. Winter
- Children's Minnesota Research Institute and Cancer and Blood Disorders ProgramChildren's MinnesotaMinneapolisMN
| | - Stephen P. Hunger
- Department of Pediatrics and the Center for Childhood Cancer ResearchChildren's Hospital of PhiladelphiaPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA
| | - Judy L. Cannon
- Department of PathologyUniversity of New Mexico Comprehensive Cancer CenterAlbuquerqueNM
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | | |
Collapse
|
49
|
Saadi W, Kermezli Y, Dao LTM, Mathieu E, Santiago-Algarra D, Manosalva I, Torres M, Belhocine M, Pradel L, Loriod B, Aribi M, Puthier D, Spicuglia S. A critical regulator of Bcl2 revealed by systematic transcript discovery of lncRNAs associated with T-cell differentiation. Sci Rep 2019; 9:4707. [PMID: 30886319 PMCID: PMC6423290 DOI: 10.1038/s41598-019-41247-5] [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] [Received: 10/08/2018] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Normal T-cell differentiation requires a complex regulatory network which supports a series of maturation steps, including lineage commitment, T-cell receptor (TCR) gene rearrangement, and thymic positive and negative selection. However, the underlying molecular mechanisms are difficult to assess due to limited T-cell models. Here we explore the use of the pro-T-cell line P5424 to study early T-cell differentiation. Stimulation of P5424 cells by the calcium ionophore ionomycin together with PMA resulted in gene regulation of T-cell differentiation and activation markers, partially mimicking the CD4-CD8- double negative (DN) to double positive (DP) transition and some aspects of subsequent T-cell maturation and activation. Global analysis of gene expression, along with kinetic experiments, revealed a significant association between the dynamic expression of coding genes and neighbor lncRNAs including many newly-discovered transcripts, thus suggesting potential co-regulation. CRISPR/Cas9-mediated genetic deletion of Robnr, an inducible lncRNA located downstream of the anti-apoptotic gene Bcl2, demonstrated a critical role of the Robnr locus in the induction of Bcl2. Thus, the pro-T-cell line P5424 is a powerful model system to characterize regulatory networks involved in early T-cell differentiation and maturation.
Collapse
Affiliation(s)
- Wiam Saadi
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Yasmina Kermezli
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Lan T M Dao
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Vinmec Research Institute of Stem cell and Gene technology (VRISG), Hanoi, Vietnam
| | - Evelyne Mathieu
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - David Santiago-Algarra
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Iris Manosalva
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Magali Torres
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Mohamed Belhocine
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.,Molecular Biology and Genetics Laboratory, Dubai, United Arab Emirates
| | - Lydie Pradel
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Beatrice Loriod
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Denis Puthier
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France. .,Equipe Labélisée Ligue Contre le Cancer, Marseille, France.
| |
Collapse
|
50
|
Cabrera JR, Manivanh R, North BJ, Leib DA. The ESCRT-Related ATPase Vps4 Is Modulated by Interferon during Herpes Simplex Virus 1 Infection. mBio 2019; 10:e02567-18. [PMID: 30837340 PMCID: PMC6401484 DOI: 10.1128/mbio.02567-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/14/2019] [Indexed: 01/03/2023] Open
Abstract
Interferons (IFNs) and autophagy are critical neuronal defenses against viral infection. IFNs alter neuronal autophagy by promoting the accumulation of IFN-dependent LC3-decorated autophagic structures, termed LC3 clusters. Here, we analyzed LC3 clusters in sensory ganglia following herpes simplex virus 1 (HSV-1) infection. In the vicinity of acutely infected neurons, antigen-negative neurons contained structures resembling accumulated autophagosomes and autolysosomes that culminated in LC3 clusters. This accumulation reflects a delayed completion of autophagy. The endosomal sorting complexes required for transport (ESCRT) machinery participates in autophagosome closure and is also required for HSV-1 replication. In this study, our results showed that HSV-1 infection in vivo and in primary neurons caused a decrease in Vps4 (a key ESCRT pathway ATPase) RNA and protein with concomitant Stat1 activation and LC3 cluster induction. We also observed that IFNs were sufficient to decrease RNA and protein levels of Vps4 in primary neurons and in other cell types. The accumulation of ubiquitin was also observed at the LC3 cluster sites. Together, our results show that IFNs modulate the ESCRT machinery in neurons in response to HSV-1 infections.IMPORTANCE Neurons rely on IFNs and autophagy as major defenses against viral infections, and HSV must overcome such defenses in order to replicate. In addition to controlling host immunity, HSV must also control host membranes in order to complete its life cycle. HSV uses the host ESCRT membrane scission machinery for viral production and transport. Here we present evidence of a new IFN-dependent mechanism used by the host to prevent ESCRT subversion by HSV. This activity also impacts the dynamics of autophagy, possibly explaining the presence of recently described LC3 clusters in the HSV-infected nervous system. The induced accumulations of ubiquitin observed in these LC3 clusters resembled those observed in certain neurodegenerative diseases, suggesting possible mechanistic parallels between these conditions.
Collapse
Affiliation(s)
- Jorge Ruben Cabrera
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire, USA
| | - Richard Manivanh
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire, USA
| | - Brian J North
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire, USA
| | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire, USA
| |
Collapse
|