1
|
Reichelt P, Bernhart S, Platzbecker U, Cross M. MicroRNA Screening Reveals Upregulation of FoxO-Signaling in Relapsed Acute Myeloid Leukemia Patients. Genes (Basel) 2024; 15:1625. [PMID: 39766892 PMCID: PMC11675194 DOI: 10.3390/genes15121625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Objectives: AML is an aggressive malignant disease characterized by aberrant proliferation and accumulation of immature blast cells in the patient's bone marrow. Chemotherapeutic treatment can effectively induce remission and re-establish functional hematopoiesis. However, many patients experience chemoresistance-associated relapse and disease progression with a poor prognosis. The identification of molecular determinants of chemoresistance that could serve as potential targets for the therapeutic restoration of chemosensitivity has proven to be challenging. Methods: To address this, we have analyzed longitudinal changes in the expression of microRNAs during disease progression in a small set of four AML patients, combined with gene ontology (GO) pathway analysis and evaluation of gene expression data in patient databases. Results: MicroRNA profiling of bone marrow samples at diagnosis and after relapse revealed significant differential expression of a large number of microRNAs between the two time points. Subsequent GO pathway analysis identified 11 signal transduction pathways likely to be affected by the differential miRNA signatures. Exemplary validation of the FoxO signaling pathway by gene expression analysis confirmed significant upregulation of FOXO1 and the target genes GADD45 and SOD2. Conclusions: Here, we show how a microRNA-based pathway prediction strategy can be used to identify differentially regulated signaling pathways that represent potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Paula Reichelt
- Department of Hematology, Cell Therapy, Hemostaseology and Infectiology, University Hospital Leipzig, 04103 Leipzig, Germany; (U.P.); (M.C.)
| | - Stephan Bernhart
- Interdisciplinary Center for Bioinformatics, Leipzig University, 04107 Leipzig, Germany;
| | - Uwe Platzbecker
- Department of Hematology, Cell Therapy, Hemostaseology and Infectiology, University Hospital Leipzig, 04103 Leipzig, Germany; (U.P.); (M.C.)
| | - Michael Cross
- Department of Hematology, Cell Therapy, Hemostaseology and Infectiology, University Hospital Leipzig, 04103 Leipzig, Germany; (U.P.); (M.C.)
| |
Collapse
|
2
|
Bruserud Ø, Reikvam H. Casein Kinase 2 (CK2): A Possible Therapeutic Target in Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:3711. [PMID: 37509370 PMCID: PMC10378128 DOI: 10.3390/cancers15143711] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The protein kinase CK2 (also known as casein kinase 2) is one of the main contributors to the human phosphoproteome. It is regarded as a possible therapeutic strategy in several malignant diseases, including acute myeloid leukemia (AML), which is an aggressive bone marrow malignancy. CK2 is an important regulator of intracellular signaling in AML cells, especially PI3K-Akt, Jak-Stat, NFκB, Wnt, and DNA repair signaling. High CK2 levels in AML cells at the first time of diagnosis are associated with decreased survival (i.e., increased risk of chemoresistant leukemia relapse) for patients receiving intensive and potentially curative antileukemic therapy. However, it is not known whether these high CK2 levels can be used as an independent prognostic biomarker because this has not been investigated in multivariate analyses. Several CK2 inhibitors have been developed, but CX-4945/silmitasertib is best characterized. This drug has antiproliferative and proapoptotic effects in primary human AML cells. The preliminary results from studies of silmitasertib in the treatment of other malignancies suggest that gastrointestinal and bone marrow toxicities are relatively common. However, clinical AML studies are not available. Taken together, the available experimental and clinical evidence suggests that the possible use of CK2 inhibition in the treatment of AML should be further investigated.
Collapse
Affiliation(s)
- Øystein Bruserud
- Institute for Clinical Science, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Institute for Clinical Science, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
3
|
Wu L, Balyan R, Brzostek J, Zhao X, Gascoigne NRJ. Time required for commitment to T cell proliferation depends on TCR affinity and cytokine response. EMBO Rep 2023; 24:e54969. [PMID: 36327141 PMCID: PMC9827553 DOI: 10.15252/embr.202254969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
T cell activation and effector functions are determined by the affinity of the interaction between T cell receptor (TCR) and its antigenic peptide MHC (pMHC) ligand. A better understanding of the quantitative aspects of TCR-pMHC affinity-dependent T cell activation is critical for the development of new immunotherapeutic strategies. However, the role of TCR-pMHC affinity in regulating the kinetics of CD8+ T cell commitment to proliferation and differentiation is unknown. Here, we show that the stronger the TCR-pMHC affinity, the shorter the time of T cell-APC co-culture required to commit CD8+ T cells to proliferation. The time threshold for T cell cytokine production is much lower than that for cell proliferation. There is a strong correlation between affinity-dependent differences in AKT phosphorylation and T cell proliferation. The cytokine IL-15 increases the poor proliferation of T cells stimulated with low affinity pMHC, suggesting that pro-inflammatory cytokines can override the affinity-dependent features of T cell proliferation.
Collapse
Affiliation(s)
- Liang‐zhe Wu
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Renu Balyan
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Present address:
Tessa Therapeutics Ltd.SingaporeSingapore
| | - Joanna Brzostek
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Present address:
Department of BiologyUniversity of FreiburgFreiburg im BreisgauGermany
| | - Xiang Zhao
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Present address:
Stanford University School of MedicineStanfordCAUSA
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
4
|
A pyridinesulfonamide derivative FD268 suppresses cell proliferation and induces apoptosis via inhibiting PI3K pathway in acute myeloid leukemia. PLoS One 2022; 17:e0277893. [PMID: 36413544 PMCID: PMC9681083 DOI: 10.1371/journal.pone.0277893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022] Open
Abstract
Aberration of PI3K signaling pathway has been confirmed to be associated with several hematological malignancies including acute myeloid leukemia (AML). FD268, a pyridinesulfonamide derivative characterized by the conjugation of 7-azaindole group, is a newly identified PI3K inhibitor showing high potent enzyme activity at nanomole concentration. In this study, we demonstrated that FD268 dose-dependently inhibits survival of AML cells with the efficacy superior to that of PI-103 (pan-PI3K inhibitor) and CAL-101 (selective PI3Kδ inhibitor) in the tested HL-60, MOLM-16, Mv-4-11, EOL-1 and KG-1 cell lines. Further mechanistic studies focused on HL-60 revealed that FD268 significantly inhibits the PI3K/Akt/mTOR signaling pathway, promotes the activation of pro-apoptotic protein Bad and downregulates the expression of anti-apoptotic protein Mcl-1, thus suppressing the cell proliferation and inducing caspase-3-dependent apoptosis. The bioinformatics analysis of the transcriptome sequencing data also indicated a potential involvement of the PI3K/Akt/mTOR pathway. These studies indicated that FD268 possesses high potent activity toward AML cells via inhibition of PI3K/Akt/mTOR signaling pathway, which sheds some light on the pyridinesulfonamide scaffold for further optimization and investigation.
Collapse
|
5
|
Wang W, Sindrewicz-Goral P, Chen C, Duckworth CA, Pritchard DM, Rhodes JM, Yu LG. Appearance of peanut agglutinin in the blood circulation after peanut ingestion promotes endothelial secretion of metastasis-promoting cytokines. Carcinogenesis 2021; 42:1079-1088. [PMID: 34223877 PMCID: PMC8643467 DOI: 10.1093/carcin/bgab059] [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: 02/03/2021] [Revised: 06/07/2021] [Accepted: 07/02/2021] [Indexed: 11/18/2022] Open
Abstract
Peanut agglutinin (PNA) is a carbohydrate-binding protein in peanuts that accounts for ~0.15% peanut weight. PNA is highly resistant to cooking and digestion and is rapidly detectable in the blood after peanut consumption. Our previous studies have shown that circulating PNA mimics the actions of endogenous galactoside-binding protein galectin-3 by interaction with tumour cell-associated MUC1 and promotes circulating tumour cell metastatic spreading. The present study shows that circulating PNA interacts with micro- as well as macro-vascular endothelial cells and induces endothelial secretion of cytokines MCP-1 (CCL2) and IL-6 in vitro and in vivo. The increased secretion of these cytokines autocrinely/paracrinely enhances the expression of endothelial cell surface adhesion molecules including integrins, VCAM and selectin, leading to increased tumour cell-endothelial adhesion and endothelial tubule formation. Binding of PNA to endothelial surface MCAM (CD146), via N-linked glycans, and subsequent activation of PI3K-AKT-PREAS40 signalling is here shown responsible for PNA-induced secretion of MCP-1 and IL-6 by vascular endothelium. Thus, in addition to its influence on promoting tumour cell spreading by interaction with tumour cell-associated MUC1, circulating PNA might also influence metastasis by enhancing the secretion of metastasis-promoting MCP-1 and IL-6 from the vascular endothelium.
Collapse
Affiliation(s)
- Weikun Wang
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Paulina Sindrewicz-Goral
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Chen Chen
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Carrie A Duckworth
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - David Mark Pritchard
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Jonathan M Rhodes
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Lu-Gang Yu
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
6
|
Aasebø E, Brenner AK, Hernandez-Valladares M, Birkeland E, Berven FS, Selheim F, Bruserud Ø. Proteomic Comparison of Bone Marrow Derived Osteoblasts and Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22115665. [PMID: 34073480 PMCID: PMC8198503 DOI: 10.3390/ijms22115665] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate into osteoblasts, and therapeutic targeting of these cells is considered both for malignant and non-malignant diseases. We analyzed global proteomic profiles for osteoblasts derived from ten and MSCs from six healthy individuals, and we quantified 5465 proteins for the osteoblasts and 5420 proteins for the MSCs. There was a large overlap in the profiles for the two cell types; 156 proteins were quantified only in osteoblasts and 111 proteins only for the MSCs. The osteoblast-specific proteins included several extracellular matrix proteins and a network including 27 proteins that influence intracellular signaling (Wnt/Notch/Bone morphogenic protein pathways) and bone mineralization. The osteoblasts and MSCs showed only minor age- and sex-dependent proteomic differences. Finally, the osteoblast and MSC proteomic profiles were altered by ex vivo culture in serum-free media. We conclude that although the proteomic profiles of osteoblasts and MSCs show many similarities, we identified several osteoblast-specific extracellular matrix proteins and an osteoblast-specific intracellular signaling network. Therapeutic targeting of these proteins will possibly have minor effects on MSCs. Furthermore, the use of ex vivo cultured osteoblasts/MSCs in clinical medicine will require careful standardization of the ex vivo handling of the cells.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (E.A.); (A.K.B.)
- Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5020 Bergen, Norway; (M.H.-V.); (E.B.); (F.S.B.); (F.S.)
| | - Annette K. Brenner
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (E.A.); (A.K.B.)
| | - Maria Hernandez-Valladares
- Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5020 Bergen, Norway; (M.H.-V.); (E.B.); (F.S.B.); (F.S.)
| | - Even Birkeland
- Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5020 Bergen, Norway; (M.H.-V.); (E.B.); (F.S.B.); (F.S.)
| | - Frode S. Berven
- Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5020 Bergen, Norway; (M.H.-V.); (E.B.); (F.S.B.); (F.S.)
| | - Frode Selheim
- Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5020 Bergen, Norway; (M.H.-V.); (E.B.); (F.S.B.); (F.S.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (E.A.); (A.K.B.)
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| |
Collapse
|
7
|
Gan D, Su Q, Su H, Wu L, Chen J, Han B, Xiang M. Burn Ointment Promotes Cutaneous Wound Healing by Modulating the PI3K/AKT/mTOR Signaling Pathway. Front Pharmacol 2021; 12:631102. [PMID: 33762951 PMCID: PMC7982805 DOI: 10.3389/fphar.2021.631102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/20/2021] [Indexed: 02/05/2023] Open
Abstract
Burn ointment (BO) is a clinically useful medicine for the treatment of burns and scalds. However, there is no enough scientific evidence to report the effect of BO on wound healing and its analgesic and anti-inflammatory efficacy. The aim of this work was to evaluate the anti-inflammatory and analgesic efficacy of BO and to reveal the potential wound healing properties and related mechanisms of BO. In this work, the content of active ingredients of BO was determined by high-performance liquid chromatography (HPLC). Two animal models of inflammation were used to study its anti-inflammatory activity, and a hot plate method was used to evaluate its analgesic effect. In addition, mouse incision and rat burn models were used to investigate the effect of BO on the anti-inflammatory and wound healing mechanisms. The results showed that BO was safe for topical application, and BO could significantly inhibit auricular swelling in mice and paw swelling in rats and significantly prolong the latency period of paw licking in the hot plate experiment in mice. It can also accelerate wound healing and repair scars by promoting the formation of new epithelial tissues in rat burn models. In addition, BO significantly downregulated the serum level of TNF-α and significantly increased the serum levels of VEGF and TGF-β1. Also, BO promoted the expression of collagen I and increased the ratio in p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR pathways. Our results demonstrate the safety and efficacy of BO and suggest that activation of the PI3K/AKT/mTOR signaling pathway may play an important role in the promotion of wound healing by BO.
Collapse
Affiliation(s)
- Dali Gan
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qiyuan Su
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Hanwen Su
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Wu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jun Chen
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Hospital of Traditional Chinese and Western Medicine), Wuhan, China
| | - Bing Han
- Department of Pathology, Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Meixian Xiang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
8
|
Daw S, Law S. Quercetin induces autophagy in myelodysplastic bone marrow including hematopoietic stem/progenitor compartment. ENVIRONMENTAL TOXICOLOGY 2021; 36:149-167. [PMID: 32902906 DOI: 10.1002/tox.23020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
Myelodysplastic syndrome (MDS) is regarded as a spectrum of bone marrow failure disorders that share hemato-pathological state of cellular dysplasia and cytopenia. The modern treatment of cancers like chemotherapy and radiation therapy sometimes severely pounce on the basic hematopoietic stem/progenitor cellular (HSPC) compartment which gradually disclose the clinical symptoms of MDS. The present study involves flowcytometric protein expression analysis of insulin growth factor receptor (IGFR), PI3K-Akt-mTOR pathway, the autophagy related proteins (ATG's), the status of antioxidative molecules SOD2 and SDF1 and apoptosis profiling in ethyl-nitroso-urea induced myelodysplasia. The redox status that is, reactive oxygen species was estimated with dihydroetidium and the status of mitochondria and lysosomes were checked by Janus green B and neutral red staining respectively, pre and post quercetin treatment in MDS bone marrow. The results revealed the activated IGFR/PI3K/Akt axis in MDS bone marrow but unconventionally both p-mTOR and autophagy (p-ATG1, p-AT6, ATG7, ATG12) was downregulated. Interestingly, post quercetin treatment an upregulation of basal autophagocytosis, reversal of oxidative damage and proper functionality of mitochondria and lysosome was recorded. Taken together, the study hinted that the PI3K-Akt-mTOR pathway does not rule over the process of autophagocytosis in HSPC's of MDS bone marrow and the isoflavanoid quercetin remarkably restored autophagocytosis and hematopoietic oxidative status toward normalcy during the progression of myelodysplasia.
Collapse
Affiliation(s)
- Suchismita Daw
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, Kolkata, West Bengal, India
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, Kolkata, West Bengal, India
| |
Collapse
|
9
|
Aasebø E, Birkeland E, Selheim F, Berven F, Brenner AK, Bruserud Ø. The Extracellular Bone Marrow Microenvironment-A Proteomic Comparison of Constitutive Protein Release by In Vitro Cultured Osteoblasts and Mesenchymal Stem Cells. Cancers (Basel) 2020; 13:cancers13010062. [PMID: 33379263 PMCID: PMC7795818 DOI: 10.3390/cancers13010062] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Normal blood cells are formed in the bone marrow by a process called hematopoiesis. This process is supported by a network of non-hematopoietic cells including connective tissue cells, blood vessel cells and bone-forming cells. However, these cells can also support the growth of cancer cells, i.e., hematological malignancies (e.g., leukemias) and cancers that arise in another organ and spread to the bone marrow. Two of these cancer-supporting normal cells are bone-forming osteoblasts and a subset of connective tissue cells called mesenchymal stem cells. One mechanism for their cancer support is the release of proteins that support cancer cell proliferation and progression of the cancer disease. Our present study shows that both these normal cells release a wide range of proteins that support cancer cells, and inhibition of this protein-mediated cancer support may become a new strategy for cancer treatment. Abstract Mesenchymal stem cells (MSCs) and osteoblasts are bone marrow stromal cells that contribute to the formation of stem cell niches and support normal hematopoiesis, leukemogenesis and development of metastases from distant cancers. This support is mediated through cell–cell contact, release of soluble mediators and formation of extracellular matrix. By using a proteomic approach, we characterized the protein release by in vitro cultured human MSCs (10 donors) and osteoblasts (nine donors). We identified 1379 molecules released by these cells, including 340 proteins belonging to the GO-term Extracellular matrix. Both cell types released a wide range of functionally heterogeneous proteins including extracellular matrix molecules (especially collagens), several enzymes and especially proteases, cytokines and soluble adhesion molecules, but also several intracellular molecules including chaperones, cytoplasmic mediators, histones and non-histone nuclear molecules. The levels of most proteins did not differ between MSCs and osteoblasts, but 82 proteins were more abundant for MSC (especially extracellular matrix proteins and proteases) and 36 proteins more abundant for osteoblasts. Finally, a large number of exosomal proteins were identified. To conclude, MSCs and osteoblasts show extracellular release of a wide range of functionally diverse proteins, including several extracellular matrix molecules known to support cancer progression (e.g., metastases from distant tumors, increased relapse risk for hematological malignancies), and the large number of identified exosomal proteins suggests that exocytosis is an important mechanism of protein release.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
| | - Even Birkeland
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Frode Selheim
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Frode Berven
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Annette K. Brenner
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
- Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
- Correspondence: or ; Tel.: +47-5597-2997
| |
Collapse
|
10
|
Li ZY, Liang C, Ding M, Weng XQ, Sheng Y, Wu J, Lu H, Cai X. Enzastaurin enhances ATRA-induced differentiation of acute myeloid leukemia cells. Am J Transl Res 2020; 12:7836-7854. [PMID: 33437364 PMCID: PMC7791522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
All-trans retinoic acid (ATRA) is considered to be the sole clinically-useful differentiating agent in the treatment of acute myeloid leukemia (AML). However, ATRA has been effective only in acute promyelocytic leukemia (APL) but not other subtypes of AML. Therefore, discovering strategies to sensitize cells to ATRA may lead to the development of ATRA-based treatments in non-APL AML patients. In the present study, a clinically-achievable concentration of enzastaurin enhanced ATRA-induced differentiation in AML cell lines, HL-60 and U937 as well as non-APL AML primary cells. Furthermore, it also restored ATRA sensitivity in ATRA-resistant cell line, HL-60Res. Mechanistically, in all these cell lines, enzastaurin-ATRA (enz-ATRA) co-treatment enhanced the protein levels of PU.1, CCAAT/enhancer-binding protein β (C/EBPβ) and C/EBPε. The activity of protein kinase C β (PKCβ) was suppressed by enz-ATRA treatment in HL-60 and HL-60Res cells. However, another PKCβ-selective inhibitor mimicked the cellular and molecular effects of enzastaurin only in HL-60 cells. Furthermore, in U937 cells, enz-ATRA activated MEK and ERK, and a MEK-specific inhibitor suppressed enz-ATRA-triggered differentiation and reduced the protein levels of PU.1, C/EBPβ and C/EBPε. Enz-ATRA activated Akt in HL-60 and HL-60Res cells. However, an Akt inhibitor blocked enz-ATRA-triggered differentiation and restored the protein levels of PU.1, C/EBPβ and C/EBPε only in HL-60Res cells. Therefore, PKCβ inhibition, MEK/ERK and Akt activation were involved in enz-ATRA-induced differentiation in HL-60, U937 and HL-60Res cells, respectively, via modulation of the protein levels of C/EBPβ, C/EBPε and PU.1. Taken together, our findings may help to guide novel therapeutic strategies for AML patients.
Collapse
Affiliation(s)
- Ze-Yi Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| | - Cui Liang
- Department of Hematology, Changhai Hospital, The Second Military Medical UniversityNo. 168 Changhai Road, Shanghai 200433, China
| | - Ming Ding
- Department of Hematology Oncology, Central Hospital of Minhang DistrictNo. 170 Xin Song Road, Shanghai 201199, China
| | - Xiang-Qin Weng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| | - Yan Sheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| | - Jing Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| | - Hao Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| | - Xun Cai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineNo. 197 Rui-jin Road II, Shanghai 200025, China
| |
Collapse
|
11
|
Targeted inhibition of cooperative mutation- and therapy-induced AKT activation in AML effectively enhances response to chemotherapy. Leukemia 2020; 35:2030-2042. [PMID: 33299144 DOI: 10.1038/s41375-020-01094-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/30/2020] [Accepted: 11/09/2020] [Indexed: 11/09/2022]
Abstract
Most AML patients exhibit mutational activation of the PI3K/AKT signaling pathway, which promotes downstream effects including growth, survival, DNA repair, and resistance to chemotherapy. Herein we demonstrate that the inv(16)/KITD816Y AML mouse model exhibits constitutive activation of PI3K/AKT signaling, which was enhanced by chemotherapy-induced DNA damage through DNA-PK-dependent AKT phosphorylation. Strikingly, inhibitors of either PI3K or DNA-PK markedly reduced chemotherapy-induced AKT phosphorylation and signaling leading to increased DNA damage and apoptosis of inv(16)/KITD816Y AML cells in response to chemotherapy. Consistently, combinations of chemotherapy and PI3K or DNA-PK inhibitors synergistically inhibited growth and survival of clonogenic AML cells without substantially inhibiting normal clonogenic bone marrow cells. Moreover, treatment of inv(16)/KITD816Y AML mice with combinations of chemotherapy and PI3K or DNA-PK inhibitors significantly prolonged survival compared to untreated/single-treated mice. Mechanistically, our findings implicate that constitutive activation of PI3K/AKT signaling driven by mutant KIT, and potentially other mutational activators such as FLT3 and RAS, cooperates with chemotherapy-induced DNA-PK-dependent activation of AKT to promote survival, DNA repair, and chemotherapy resistance in AML. Hence, our study provides a rationale to select AML patients exhibiting constitutive PI3K/AKT activation for simultaneous treatment with chemotherapy and inhibitors of DNA-PK and PI3K to improve chemotherapy response and clinical outcome.
Collapse
|
12
|
Annageldiyev C, Tan SF, Thakur S, Dhanyamraju PK, Ramisetti SR, Bhadauria P, Schick J, Zeng Z, Sharma V, Dunton W, Dovat S, Desai D, Zheng H, Feith DJ, Loughran TP, Amin S, Sharma AK, Claxton D, Sharma A. The PI3K/AKT Pathway Inhibitor ISC-4 Induces Apoptosis and Inhibits Growth of Leukemia in Preclinical Models of Acute Myeloid Leukemia. Front Oncol 2020; 10:393. [PMID: 32296637 PMCID: PMC7140985 DOI: 10.3389/fonc.2020.00393] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia is a heterogeneous disease with a 5-year survival rate of 28.3%, and current treatment options constrained by dose-limiting toxicities. One of the key signaling pathways known to be frequently activated and dysregulated in AML is PI3K/AKT. Its dysregulation is associated with aggressive cell growth and drug resistance. We investigated the activity of Phenybutyl isoselenocyanate (ISC-4) in primary cells obtained from newly diagnosed AML patients, diverse AML cell lines, and normal cord blood cells. ISC-4 significantly inhibited survival and clonogenicity of primary human AML cells without affecting normal cells. We demonstrated that ISC-4-mediated p-Akt inhibition caused apoptosis in primary AML (CD34+) stem cells and enhanced efficacy of cytarabine. ISC-4 impeded leukemia progression with improved overall survival in a syngeneic C1498 mouse model with no obvious toxic effects on normal myelopoiesis. In U937 xenograft model, bone marrow cells exhibited significant reduction in human CD45+ cells in ISC-4 (~87%) or AraC (~89%) monotherapy groups compared to control. Notably, combination treatment suppressed the leukemic infiltration significantly higher than the single-drug treatments (~94%). Together, the present findings suggest that ISC-4 might be a promising agent for AML treatment.
Collapse
Affiliation(s)
- Charyguly Annageldiyev
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Su-Fern Tan
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Shreya Thakur
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Pavan Kumar Dhanyamraju
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Srinivasa R Ramisetti
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Preeti Bhadauria
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Jacob Schick
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Zheng Zeng
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Varun Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Wendy Dunton
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Dhimant Desai
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Hong Zheng
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David J Feith
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Thomas P Loughran
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Shantu Amin
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arun K Sharma
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arati Sharma
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
13
|
Tarantelli C, Lupia A, Stathis A, Bertoni F. Is There a Role for Dual PI3K/mTOR Inhibitors for Patients Affected with Lymphoma? Int J Mol Sci 2020; 21:E1060. [PMID: 32033478 PMCID: PMC7037719 DOI: 10.3390/ijms21031060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The activation of the PI3K/AKT/mTOR pathway is a main driver of cell growth, proliferation, survival, and chemoresistance of cancer cells, and, for this reason, represents an attractive target for developing targeted anti-cancer drugs. There are plenty of preclinical data sustaining the anti-tumor activity of dual PI3K/mTOR inhibitors as single agents and in combination in lymphomas. Clinical responses, including complete remissions (especially in follicular lymphoma patients), are also observed in the very few clinical studies performed in patients that are affected by relapsed/refractory lymphomas or chronic lymphocytic leukemia. In this review, we summarize the literature on dual PI3K/mTOR inhibitors focusing on the lymphoma setting, presenting both the three compounds still in clinical development and those with a clinical program stopped or put on hold.
Collapse
Affiliation(s)
- Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
| | - Antonio Lupia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, USI, 6900 Lugano, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
| |
Collapse
|
14
|
Leufven E, Bruserud Ø. Immunosuppression and Immunotargeted Therapy in Acute Myeloid Leukemia - The Potential Use of Checkpoint Inhibitors in Combination with Other Treatments. Curr Med Chem 2019; 26:5244-5261. [PMID: 30907305 DOI: 10.2174/0929867326666190325095853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Immunotherapy by using checkpoint inhibitors is now tried in the treatment of several malignancies, including Acute Myeloid Leukemia (AML). The treatment is tried both as monotherapy and as a part of combined therapy. METHODS Relevant publications were identified through literature searches in the PubMed database. We searched for (i) original articles describing the results from clinical studies of checkpoint inhibition; (ii) published articles describing the immunocompromised status of AML patients; and (iii) published studies of antileukemic immune reactivity and immunotherapy in AML. RESULTS Studies of monotherapy suggest that checkpoint inhibition has a modest antileukemic effect and complete hematological remissions are uncommon, whereas combination with conventional chemotherapy increases the antileukemic efficiency with acceptable toxicity. The experience with a combination of different checkpoint inhibitors is limited. Thalidomide derivatives are referred to as immunomodulatory drugs and seem to reverse leukemia-induced immunosuppression, but in addition, they have direct inhibitory effects on the AML cells. The combination of checkpoint targeting and thalidomide derivatives thus represents a strategy for dual immunotargeting together with a direct antileukemic effect. CONCLUSION Checkpoint inhibitors are now tried in AML. Experimental studies suggest that these inhibitors should be combined with immunomodulatory agents (i.e. thalidomide derivatives) and/or new targeted or conventional antileukemic treatment. Such combinations would allow dual immunotargeting (checkpoint inhibitor, immunomodulatory agents) together with a double/triple direct targeting of the leukemic cells.
Collapse
Affiliation(s)
- Eva Leufven
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway.,Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021, Bergen, Norway
| |
Collapse
|
15
|
Lv Y, Tan J, Miao Y, Zhang Q. The role of microvesicles and its active molecules in regulating cellular biology. J Cell Mol Med 2019; 23:7894-7904. [PMID: 31559684 PMCID: PMC6850934 DOI: 10.1111/jcmm.14667] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 12/17/2022] Open
Abstract
Cell‐derived microvesicles are membrane vesicles produced by the outward budding of the plasma membrane and released by almost all types of cells. These have been considered as another mechanism of intercellular communication, because they carry active molecules, such as proteins, lipids and nucleic acids. Furthermore, these are present in circulating fluids, such as blood and urine, and are closely correlated to the progression of pathophysiological conditions in many diseases. Recent studies have revealed that microvesicles have a dual effect of damage and protection of receptor cells. However, the nature of the active molecules involved in this effect remains unclear. The present study mainly emphasized the mechanism of microvesicles and the active molecules mediating the different biological effects of receptor cells by affecting autophagy, apoptosis and inflammation pathways. The effective ways of blocking microvesicles and its active molecules in mediating cell damage when microvesicles exert harmful effects were also discussed.
Collapse
Affiliation(s)
- YingMei Lv
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
16
|
Brattås MK, Reikvam H, Tvedt THA, Bruserud Ø. Precision medicine for TP53-mutated acute myeloid leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1644164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
High Constitutive Cytokine Release by Primary Human Acute Myeloid Leukemia Cells Is Associated with a Specific Intercellular Communication Phenotype. J Clin Med 2019; 8:jcm8070970. [PMID: 31277464 PMCID: PMC6678419 DOI: 10.3390/jcm8070970] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease, and this heterogeneity includes the capacity of constitutive release of extracellular soluble mediators by AML cells. We investigated whether this capacity is associated with molecular genetic abnormalities, and we compared the proteomic profiles of AML cells with high and low release. AML cells were derived from 71 consecutive patients that showed an expected frequency of cytogenetic and molecular genetic abnormalities. The constitutive extracellular release of 34 soluble mediators (CCL and CXCL chemokines, interleukins, proteases, and protease regulators) was investigated for an unselected subset of 62 patients, and they could be classified into high/intermediate/low release subsets based on their general capacity of constitutive secretion. FLT3-ITD was more frequent among patients with high constitutive mediator release, but our present study showed no additional associations between the capacity of constitutive release and 53 other molecular genetic abnormalities. We compared the proteomic profiles of two contrasting patient subsets showing either generally high or low constitutive release. A network analysis among cells with high release levels demonstrated high expression of intracellular proteins interacting with integrins, RAC1, and SYK signaling. In contrast, cells with low release showed high expression of several transcriptional regulators. We conclude that AML cell capacity of constitutive mediator release is characterized by different expression of potential intracellular therapeutic targets.
Collapse
|
18
|
Nepstad I, Hatfield KJ, Grønningsæter IS, Aasebø E, Hernandez-Valladares M, Hagen KM, Rye KP, Berven FS, Selheim F, Reikvam H, Bruserud Ø. Effects of insulin and pathway inhibitors on the PI3K-Akt-mTOR phosphorylation profile in acute myeloid leukemia cells. Signal Transduct Target Ther 2019; 4:20. [PMID: 31240133 PMCID: PMC6582141 DOI: 10.1038/s41392-019-0050-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/05/2019] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)-Akt-mechanistic target of rapamycin (mTOR) pathway is constitutively activated in human acute myeloid leukemia (AML) cells and is regarded as a possible therapeutic target. Insulin is an agonist of this pathway and a growth factor for AML cells. We characterized the effect of insulin on the phosphorylation of 10 mediators in the main track of the PI3K-Akt-mTOR pathway in AML cells from 76 consecutive patients. The overall results showed that insulin significantly increased the phosphorylation of all investigated mediators. However, insulin effects on the pathway activation profile varied among patients, and increased phosphorylation in all mediators was observed only in a minority of patients; in other patients, insulin had divergent effects. Global gene expression profiling and proteomic/phosphoproteomic comparisons suggested that AML cells from these two patient subsets differed with regard to AML cell differentiation, transcriptional regulation, RNA metabolism, and cellular metabolism. Strong insulin-induced phosphorylation was associated with weakened antiproliferative effects of metabolic inhibitors. PI3K, Akt, and mTOR inhibitors also caused divergent effects on the overall pathway phosphorylation profile in the presence of insulin, although PI3K and Akt inhibition caused a general reduction in Akt pT308 and 4EBP1 pT36/pT45 phosphorylation. For Akt inhibition, the phosphorylation of upstream mediators was generally increased or unaltered. In contrast, mTOR inhibition reduced mTOR pS2448 and S6 pS244 phosphorylation but increased Akt pT308 phosphorylation. In conclusion, the effects of both insulin and PI3K-Akt-mTOR inhibitors differ between AML patient subsets, and differences in insulin responsiveness are associated with differential susceptibility to metabolic targeting.
Collapse
Affiliation(s)
- Ina Nepstad
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kimberley Joanne Hatfield
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ida Sofie Grønningsæter
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elise Aasebø
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Maria Hernandez-Valladares
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Karen Marie Hagen
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin Paulsen Rye
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Frode S. Berven
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Frode Selheim
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Håkon Reikvam
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
19
|
Li J, Lu L, Zhang YH, Xu Y, Liu M, Feng K, Chen L, Kong X, Huang T, Cai YD. Identification of leukemia stem cell expression signatures through Monte Carlo feature selection strategy and support vector machine. Cancer Gene Ther 2019; 27:56-69. [PMID: 31138902 DOI: 10.1038/s41417-019-0105-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/28/2019] [Accepted: 05/04/2019] [Indexed: 01/09/2023]
Abstract
Acute myeloid leukemia (AML) is a type of blood cancer characterized by the rapid growth of immature white blood cells from the bone marrow. Therapy resistance resulting from the persistence of leukemia stem cells (LSCs) are found in numerous patients. Comparative transcriptome studies have been previously conducted to analyze differentially expressed genes between LSC+ and LSC- cells. However, these studies mainly focused on a limited number of genes with the most obvious expression differences between the two cell types. We developed a computational approach incorporating several machine learning algorithms, including Monte Carlo feature selection (MCFS), incremental feature selection (IFS), support vector machine (SVM), Repeated Incremental Pruning to Produce Error Reduction (RIPPER), to identify gene expression features specific to LSCs. One thousand 0ne hudred fifty-nine features (genes) were first identified, which can be used to build the optimal SVM classifier for distinguishing LSC+ and LSC- cells. Among these 1159 genes, the top 17 genes were identified as LSC-specific biomarkers. In addition, six classification rules were produced by RIPPER algorithm. The subsequent literature review on these features/genes and the classification rules and functional enrichment analyses of the 1159 features/genes confirmed the relevance of extracted genes and rules to the characteristics of LSCs.
Collapse
Affiliation(s)
- JiaRui Li
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.,School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Yu-Hang Zhang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - Min Liu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, P. R. China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic, Guangzhou, 510507, P. R. China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, P. R. China.,Shanghai Key Laboratory of PMMP, East China Normal University, Shanghai, 200241, P. R. China
| | - XiangYin Kong
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
20
|
Abstract
Acute myeloid leukemia (AML) is a kind of malignant hematopoietic system disease characterized by abnormal proliferation, poor cell differentiation, and infiltration of bone marrow, peripheral blood, or other tissues. To date, the first-line treatment of AML is still based on daunorubicin and cytosine arabinoside or idarubicin and cytosine arabinoside regimen. However, the complete remission rate of AML is still not optimistic, especially in elderly patients, and the recurrence rate after complete remission is still high. The resistance of leukemia cells to chemotherapy drugs becomes the main obstacle in the treatment of AML. At present, the research on the mechanisms of drug resistance in AML is very active. This article will elaborate on the main mechanisms of drug resistance currently being studied, including drug resistance-related proteins and enzymes, gene alterations, micro RNAs, and signal pathways.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, People's Republic of China,
| | - Yan Gu
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, People's Republic of China,
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, People's Republic of China,
| |
Collapse
|
21
|
Guo N, Azadniv M, Coppage M, Nemer M, Mendler J, Becker M, Liesveld J. Effects of Neddylation and mTOR Inhibition in Acute Myelogenous Leukemia. Transl Oncol 2019; 12:602-613. [PMID: 30699367 PMCID: PMC6348338 DOI: 10.1016/j.tranon.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 01/26/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a heterogeneous disease and often relapses after standard chemotherapy. Recently, the neddylation (NEDD8) and the mammalian target of rapamycin (mTOR) signaling pathways have emerged as promising pharmaceutical targets for AML therapy. However, the interaction of these two pathways remains unclear. Here we evaluated the effects of pevonedistat, an inhibitor of the NEDD8 activating enzyme (NAE), and sapanisertib (TAK-228), an inhibitor of mTORC1 and mTORC2 as single agents or in combination on AML cell lines. We found that inhibition of neddylation with pevonedistat partially inhibited mTOR signaling transduction and vice versa, inhibition of mTOR signaling with sapanisertib partially inhibited neddylation in AML cell lines. Pevonedistat alone was able to induce cytotoxicity in most AML cell lines as well as in primary AML, whereas sapanisertib alone decreased cell metabolic activity, reduced cell size and arrested cells in G0 phase with only minimal induction of cell death. In addition, pevonedistat was able to induce cell differentiation, arrest cells in G2/M cell cycle phases, and induce DNA re-replication and damage. However, co-treatment with sapanisertib suppressed pevonedistat induced apoptosis, differentiation, S/G2/M arrest, and DNA damage. Taken together, our data demonstrate that pevonedistat and sapanisertib exhibit distinct anti-tumor effects on AML cells, i.e. cytotoxic and cytostatic effects, respectively; however, sapanisertib can attenuate pevonedistat-induced cellular responses in AML cells. Understanding mTOR and neddylation pathway interaction could provide therapeutic strategies for treatment of AML and other malignancies.
Collapse
Affiliation(s)
- Naxin Guo
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Mitra Azadniv
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Myra Coppage
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Mary Nemer
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Jason Mendler
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642; Department of Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Michael Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642; Department of Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642
| | - Jane Liesveld
- Wilmot Cancer Institute, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642; Department of Medicine, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642.
| |
Collapse
|
22
|
Mentoor I, Engelbrecht AM, Nell T. Fatty acids: Adiposity and breast cancer chemotherapy, a bad synergy? Prostaglandins Leukot Essent Fatty Acids 2019; 140:18-33. [PMID: 30553399 DOI: 10.1016/j.plefa.2018.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Globally, breast cancer continues to be a major concern in women's health. Lifestyle related risk factors, specifically excess adipose tissue (adiposity) has reached epidemic proportions and has been identified as a major risk factor in the development of breast cancer. Dysfunctional adipose tissue has evoked research focusing on its association with metabolic-related conditions, breast cancer risk and progression. Adipose dysfunction in coordination with immune cells and inflammation, are responsible for accelerated cell growth and survival of cancer cells. Recently, evidence also implicates adiposity as a potential risk factor for chemotherapy resistance. Chemotherapeutic agents have been shown to negatively impact adipose tissue. Since adipose tissue is a major storage site for fatty acids, it is not unlikely that these negative effects may disrupt adipose tissue homeostasis. It is therefore argued that fatty acid composition may be altered due to the chemotherapeutic pharmacokinetics, which in turn could have severe health related outcomes. The underlying molecular mechanisms elucidating the effects of fatty acid composition in adiposity-linked drug resistance are still unclear and under explored. This review focuses on the potential role of adiposity in breast cancer and specifically emphasizes the role of fatty acids in cancer progression and treatment resistance.
Collapse
Affiliation(s)
- Ilze Mentoor
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - A-M Engelbrecht
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - Theo Nell
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa.
| |
Collapse
|
23
|
Halu A, Wang JG, Iwata H, Mojcher A, Abib AL, Singh SA, Aikawa M, Sharma A. Context-enriched interactome powered by proteomics helps the identification of novel regulators of macrophage activation. eLife 2018; 7:37059. [PMID: 30303482 PMCID: PMC6179386 DOI: 10.7554/elife.37059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
The role of pro-inflammatory macrophage activation in cardiovascular disease (CVD) is a complex one amenable to network approaches. While an indispensible tool for elucidating the molecular underpinnings of complex diseases including CVD, the interactome is limited in its utility as it is not specific to any cell type, experimental condition or disease state. We introduced context-specificity to the interactome by combining it with co-abundance networks derived from unbiased proteomics measurements from activated macrophage-like cells. Each macrophage phenotype contributed to certain regions of the interactome. Using a network proximity-based prioritization method on the combined network, we predicted potential regulators of macrophage activation. Prediction performance significantly increased with the addition of co-abundance edges, and the prioritized candidates captured inflammation, immunity and CVD signatures. Integrating the novel network topology with transcriptomics and proteomics revealed top candidate drivers of inflammation. In vitro loss-of-function experiments demonstrated the regulatory role of these proteins in pro-inflammatory signaling. When human cells or tissues are injured, the body triggers a response known as inflammation to repair the damage and protect itself from further harm. However, if the same issue keeps recurring, the tissues become inflamed for longer periods of time, which may ultimately lead to health problems. This is what could be happening in cardiovascular diseases, where long-term inflammation could damage the heart and blood vessels. Many different proteins interact with each other to control inflammation; gaining an insight into the nature of these interactions could help to pinpoint the role of each molecular actor. Researchers have used a combination of unbiased, large-scale experimental and computational approaches to develop the interactome, a map of the known interactions between all proteins in humans. However, interactions between proteins can change between cell types, or during disease. Here, Halu et al. aimed to refine the human interactome and identify new proteins involved in inflammation, especially in the context of cardiovascular disease. Cells called macrophages produce signals that trigger inflammation whey they detect damage in other cells or tissues. The experiments used a technique called proteomics to measure the amounts of all the proteins in human macrophages. Combining these data with the human interactome made it possible to predict new links between proteins known to have a role in inflammation and other proteins in the interactome. Further analysis using other sets of data from macrophages helped identify two new candidate proteins – GBP1 and WARS – that may promote inflammation. Halu et al. then used a genetic approach to deactivate the genes and decrease the levels of these two proteins in macrophages, which caused the signals that encourage inflammation to drop. These findings suggest that GBP1 and WARS regulate the activity of macrophages to promote inflammation. The two proteins could therefore be used as drug targets to treat cardiovascular diseases and other disorders linked to inflammation, but further studies will be needed to precisely dissect how GBP1 and WARS work in humans.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States.,Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Jian-Guo Wang
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Alexander Mojcher
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Ana Luisa Abib
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
24
|
Clonal Heterogeneity Reflected by PI3K-AKT-mTOR Signaling in Human Acute Myeloid Leukemia Cells and Its Association with Adverse Prognosis. Cancers (Basel) 2018; 10:cancers10090332. [PMID: 30223538 PMCID: PMC6162751 DOI: 10.3390/cancers10090332] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/05/2018] [Accepted: 09/13/2018] [Indexed: 01/01/2023] Open
Abstract
Clonal heterogeneity detected by karyotyping is a biomarker associated with adverse prognosis in acute myeloid leukemia (AML). Constitutive activation of the phosphatidylinositol-3-kinase-Akt-mechanistic target of rapamycin (PI3K-Akt-mTOR) pathway is present in AML cells, and this pathway integrates signaling from several upstream receptors/mediators. We suggest that this pathway reflects biologically important clonal heterogeneity. We investigated constitutive PI3K-Akt-mTOR pathway activation in primary human AML cells derived from 114 patients, together with 18 pathway mediators. The cohort included patients with normal karyotype or single karyotype abnormalities and with an expected heterogeneity of molecular genetic abnormalities. Clonal heterogeneity reflected as pathway mediator heterogeneity was detected for 49 patients. Global gene expression profiles of AML cell populations with and without clonal heterogeneity differed with regard to expression of ectopic olfactory receptors (a subset of G-protein coupled receptors) and proteins involved in G-protein coupled receptor signaling. Finally, the presence of clonal heterogeneity was associated with adverse prognosis for patients receiving intensive antileukemic treatment. The clonal heterogeneity as reflected in the activation status of selected mediators in the PI3K-Akt-mTOR pathway was associated with a different gene expression profile and had an independent prognostic impact. Biological heterogeneity reflected in the intracellular signaling status should be further investigated as a prognostic biomarker in human AML.
Collapse
|
25
|
Biological Aspects of mTOR in Leukemia. Int J Mol Sci 2018; 19:ijms19082396. [PMID: 30110936 PMCID: PMC6121663 DOI: 10.3390/ijms19082396] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a central processor of intra- and extracellular signals, regulating many fundamental cellular processes such as metabolism, growth, proliferation, and survival. Strong evidences have indicated that mTOR dysregulation is deeply implicated in leukemogenesis. This has led to growing interest in the development of modulators of its activity for leukemia treatment. This review intends to provide an outline of the principal biological and molecular functions of mTOR. We summarize the current understanding of how mTOR interacts with microRNAs, with components of cell metabolism, and with controllers of apoptotic machinery. Lastly, from a clinical/translational perspective, we recapitulate the therapeutic results in leukemia, obtained by using mTOR inhibitors as single agents and in combination with other compounds.
Collapse
|
26
|
Nepstad I, Hatfield KJ, Aasebø E, Hernandez-Valladares M, Brenner AK, Bartaula-Brevik S, Berven F, Selheim F, Skavland J, Gjertsen BT, Reikvam H, Bruserud Ø. Two acute myeloid leukemia patient subsets are identified based on the constitutive PI3K-Akt-mTOR signaling of their leukemic cells; a functional, proteomic, and transcriptomic comparison. Expert Opin Ther Targets 2018; 22:639-653. [DOI: 10.1080/14728222.2018.1487401] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ina Nepstad
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Kimberley J. Hatfield
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Elise Aasebø
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | | | - Annette K. Brenner
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | | | - Frode Berven
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Frode Selheim
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Jørn Skavland
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Bjørn Tore Gjertsen
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
| | - Håkon Reikvam
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Section for Hematology, Department of Clinical Science, University of Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
27
|
Bartaula-Brevik S, Lindstad Brattås MK, Tvedt THA, Reikvam H, Bruserud Ø. Splenic tyrosine kinase (SYK) inhibitors and their possible use in acute myeloid leukemia. Expert Opin Investig Drugs 2018; 27:377-387. [DOI: 10.1080/13543784.2018.1459562] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Sushma Bartaula-Brevik
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Tor Henrik Anderson Tvedt
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Håkon Reikvam
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
28
|
Resistance to the Antiproliferative In Vitro Effect of PI3K-Akt-mTOR Inhibition in Primary Human Acute Myeloid Leukemia Cells Is Associated with Altered Cell Metabolism. Int J Mol Sci 2018; 19:ijms19020382. [PMID: 29382066 PMCID: PMC5855604 DOI: 10.3390/ijms19020382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/17/2018] [Accepted: 01/23/2018] [Indexed: 12/02/2022] Open
Abstract
Constitutive signaling through the phosphatidylinositol-3-kinase-Akt-mechanistic target of rapamycin (PI3K-Akt-mTOR) pathway is present in acute myeloid leukemia (AML) cells. However, AML is a heterogeneous disease, and we therefore investigated possible associations between cellular metabolism and sensitivity to PI3K-Akt-mTOR pathway inhibitors. We performed non-targeted metabolite profiling to compare the metabolome differences of primary human AML cells derived from patients susceptible or resistant to the in vitro antiproliferative effects of mTOR and PI3K inhibitors. In addition, the phosphorylation status of 18 proteins involved in PI3K-Akt-mTOR signaling and the effect of the cyclooxygenase inhibitor indomethacin on their phosphorylation status was investigated by flow cytometry. Strong antiproliferative effects by inhibitors were observed only for a subset of patients. We compared the metabolite profiles for responders and non-responders towards PI3K-mTOR inhibitors, and 627 metabolites could be detected. Of these metabolites, 128 were annotated and 15 of the annotated metabolites differed significantly between responders and non-responders, including metabolites involved in energy, amino acid, and lipid metabolism. To conclude, leukemia cells that are susceptible or resistant to PI3K-Akt-mTOR inhibitors differ in energy, amino acid, and arachidonic acid metabolism, and modulation of arachidonic acid metabolism alters the activation of mTOR and its downstream mediators.
Collapse
|
29
|
Wan L, Tian Y, Zhang R, Peng Z, Sun J, Zhang W. MicroRNA-103 confers the resistance to long-treatment of adriamycin to human leukemia cells by regulation of COP1. J Cell Biochem 2018; 119:3843-3852. [PMID: 29058777 DOI: 10.1002/jcb.26431] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
Adriamycin (ADR) is an anti-cancer drug which offers improvement in survival for acute myeloid leukemia (AML) patients. However, the drug resistance is almost inevitable. Increasing evidences suggested that microRNAs (miRNAs) were associated with cancer chemo-resistance. Here, we aimed to explore the possible mechanism of miR-103 affected resistance to ADR in AML cells. Different concentrations of ADR were used to induce K562 and KASUMI-1 cells, and miR-103 mimic, inhibitor were transfected into K562 and KASUMI-1 cells. Cell viability and proliferation were determined by trypan blue staining and MTT assays for evaluating K562 and KASUMI-1 cells drug resistance. The relationship of miR-103 and COP1, Trib1, and C/EBPα were analyzed by qRT-PCR and Western blot. Cell proliferation, viability were detected again. Besides, the expressions of main factors of cell cycle and PI3K/AKT signal pathway were analyzed by Western blot. Results showed that ADR inhibited cell viability and proliferation in K562 and KASUMI-1 cells. However, K562 and KASUMI-1 cells appeared drug resistance for 50 passages at 0.8 µM of ADR. In addition, miR-103 expression was up-regulated in ADR-resistant K562 cells (K562/ADR) and overexpression of miR-103 increased K562 cells drug resistance via promoting cell viability and cell cycle-related factors expressions. COP1 was positively regulated by miR-103, suppression of miR-103 recovered K562/ADR cells drug resistance by regulation of COP1, Trib1, and C/EBPα. Besides, miR-103 blocked PI3K/AKT signal pathway by regulation of COP1. These data indicated that miR-103 was up-regulated in drug resistant cells and it may regulate ADR-resistance by regulation of COP1 in AML cells.
Collapse
Affiliation(s)
- Lin Wan
- Emergency Department, Hospital of Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yanlong Tian
- Department of Pathology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, China
| | - Rui Zhang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhuo Peng
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiangli Sun
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
30
|
Herschbein L, Liesveld JL. Dueling for dual inhibition: Means to enhance effectiveness of PI3K/Akt/mTOR inhibitors in AML. Blood Rev 2017; 32:235-248. [PMID: 29276026 DOI: 10.1016/j.blre.2017.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/10/2017] [Accepted: 11/30/2017] [Indexed: 01/04/2023]
Abstract
The phosphatidylinositol 3-kinase/protein kinase B (Akt)/mechanistic target of rapamycin (PI3K/Akt/mTOR) pathway is amplified in 60-80% of patients with acute myelogenous leukemia (AML). Since this complex pathway is crucial to cell functions such as growth, proliferation, and survival, inhibition of this pathway would be postulated to inhibit leukemia initiation and propagation. Inhibition of the mTORC1 pathway has met with limited success in AML due to multiple resistance mechanisms including direct insensitivity of the mTORC1 complex, feedback activation of the PI3k/Akt signaling network, insulin growth factor-1 (IGF-1) activation of PI3K, and others. This review explores the role of mTOR inhibition in AML, mechanisms of resistance, and means to improve outcomes through use of dual mTORC1/2 inhibitors or dual TORC/PI3K inhibitors. How these inhibitors interface with currently available therapies in AML will require additional preclinical experiments and conduct of well-designed clinical trials.
Collapse
Affiliation(s)
- Lauren Herschbein
- Department of Medicine, The James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA.
| | - Jane L Liesveld
- Department of Medicine, The James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
31
|
A molecular signature of dormancy in CD34 +CD38 - acute myeloid leukaemia cells. Oncotarget 2017; 8:111405-111418. [PMID: 29340063 PMCID: PMC5762331 DOI: 10.18632/oncotarget.22808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/14/2017] [Indexed: 01/11/2023] Open
Abstract
Dormant leukaemia initiating cells in the bone marrow niche are a crucial therapeutic target for total eradication of acute myeloid leukaemia. To study this cellular subset we created and validated an in vitro model employing the cell line TF-1a, treated with Transforming Growth Factor β1 (TGFβ1) and a mammalian target of rapamycin inhibitor. The treated cells showed decreases in total RNA, Ki-67 and CD71, increased aldehyde dehydrogenase activity, forkhead box 03A (FOX03A) nuclear translocation and growth inhibition, with no evidence of apoptosis or differentiation. Using human genome gene expression profiling we identified a signature enriched for genes involved in adhesion, stemness/inhibition of differentiation and tumour suppression as well as canonical cell cycle regulation. The most upregulated gene was the osteopontin-coding gene SPP1. Dormant cells also demonstrated significantly upregulated beta 3 integrin (ITGB3) and CD44, as well as increased adhesion to their ligands vitronectin and hyaluronic acid as well as to bone marrow stromal cells. Immunocytochemistry of bone marrow biopsies of AML patients confirmed the positive expression of osteopontin in blasts near the para-trabecular bone marrow, whereas osteopontin was rarely detected in mononuclear cell isolates. Unsupervised hierarchical clustering of the dormancy gene signature in primary acute myeloid leukaemia samples from the Cancer Genome Atlas identified a cluster enriched for dormancy genes associated with poor overall survival.
Collapse
|
32
|
Aasebø E, Bartaula-Brevik S, Hernandez-Valladares M, Bruserud Ø. Vacuolar ATPase as a possible therapeutic target in human acute myeloid leukemia. Expert Rev Hematol 2017; 11:13-24. [PMID: 29168399 DOI: 10.1080/17474086.2018.1407239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION V-ATPase is a proton pump expressed both in the membrane of intracellular organelles (e.g. endosomes, lysosomes, Golgi structures) and the plasma membrane. It is an important regulator of organellar functions, intracellular molecular trafficking, intercellular communication and intracellular signaling. It is therefore considered as a possible therapeutic target in the treatment of human malignancies. Areas covered: Relevant publications were identified through literature searches in the PubMed database. We searched for original articles and reviews describing the possible importance of V-ATPase for leukemogenesis and chemosensitivity in human myeloid cells, especially acute myeloid leukemia (AML) cells. Expert commentary: The expression of V-ATPase in the primary human AML cells varies between patients, and high levels are associated with high constitutive release of a wide range of soluble mediators. Several of the molecules included in the V-ATPase interactome may also be important in leukemogenesis and/or development of chemoresistance in human AML. Therapeutic targeting of V-ATPase should therefore be regarded as a possible therapeutic strategy in human AML, but the efficiency of such targeting will probably differ between patients. The possibility of toxicity, especially hematological toxicity and immunosuppression, also has to be clarified.
Collapse
Affiliation(s)
- Elise Aasebø
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Sushma Bartaula-Brevik
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Maria Hernandez-Valladares
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Øystein Bruserud
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,c Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
33
|
Bruserud Ø, Aasebø E, Hernandez-Valladares M, Tsykunova G, Reikvam H. Therapeutic targeting of leukemic stem cells in acute myeloid leukemia - the biological background for possible strategies. Expert Opin Drug Discov 2017; 12:1053-1065. [PMID: 28748730 DOI: 10.1080/17460441.2017.1356818] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is an aggressive malignancy, caused by the accumulation of immature leukemic blasts in blood and bone marrow. There is a relatively high risk of chemoresistant relapse even for the younger patients who can receive the most intensive antileukemic treatment. Treatment directed against the remaining leukemic and preleukemic stem cells will most likely reduce the risk of later relapse. Areas covered: Relevant publications were identified through literature searches. The authors searched for original articles and recent reviews describing (i) the characteristics of leukemic/preleukemic stem cells; (ii) the importance of the bone marrow stem cell niches in leukemogenesis; and (iii) possible therapeutic strategies to target the preleukemic/leukemic stem cells. Expert opinion: Leukemia relapse/progression seems to be derived from residual chemoresistant leukemic or preleukemic stem cells, and a more effective treatment directed against these cells will likely be important to improve survival both for patients receiving intensive treatment and leukemia-stabilizing therapy. Several possible strategies are now considered, including the targeting of the epigenetic regulation of gene expression, proapoptotic intracellular signaling, cell metabolism, telomere activity and the AML-supporting effects by neighboring stromal cells. Due to disease heterogeneity, the most effective stem cell-directed therapy will probably differ between individual patients.
Collapse
Affiliation(s)
- Øystein Bruserud
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Elise Aasebø
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,c Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Maria Hernandez-Valladares
- a Division of Hematology, Institute of Clinical Science , University of Bergen , Bergen , Norway.,c Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Galina Tsykunova
- b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| | - Håkon Reikvam
- b Section of Hematology, Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
34
|
Hujber Z, Petővári G, Szoboszlai N, Dankó T, Nagy N, Kriston C, Krencz I, Paku S, Ozohanics O, Drahos L, Jeney A, Sebestyén A. Rapamycin (mTORC1 inhibitor) reduces the production of lactate and 2-hydroxyglutarate oncometabolites in IDH1 mutant fibrosarcoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:74. [PMID: 28578659 PMCID: PMC5457553 DOI: 10.1186/s13046-017-0544-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Multiple studies concluded that oncometabolites (e.g. D-2-hydroxyglutarate (2-HG) related to mutant isocitrate dehydrogenase 1/2 (IDH1/2) and lactate) have tumour promoting potential. Regulatory mechanisms implicated in the maintenance of oncometabolite production have great interest. mTOR (mammalian target of rapamycin) orchestrates different pathways, influences cellular growth and metabolism. Considering hyperactivation of mTOR in several malignancies, the question has been addressed whether mTOR operates through controlling of oncometabolite accumulation in metabolic reprogramming. METHODS HT-1080 cells - carrying originally endogenous IDH1 mutation - were used in vitro and in vivo. Anti-tumour effects of rapamycin were studied using different assays. The main sources and productions of the oncometabolites (2-HG and lactate) were analysed by 13C-labeled substrates. Alterations at protein and metabolite levels were followed by Western blot, flow cytometry, immunohistochemistry and liquid chromatography mass spectrometry using rapamycin, PP242 and different glutaminase inhibitors, as well. RESULTS Rapamycin (mTORC1 inhibitor) inhibited proliferation, migration and altered the metabolic activity of IDH1 mutant HT-1080 cells. Rapamycin reduced the level of 2-HG sourced mainly from glutamine and glucose derived lactate which correlated to the decreased incorporation of 13C atoms from 13C-substrates. Additionally, decreased expressions of lactate dehydrogenase A and glutaminase were also observed both in vitro and in vivo. CONCLUSIONS Considering the role of lactate and 2-HG in regulatory network and in metabolic symbiosis it could be assumed that mTOR inhibitors have additional effects besides their anti-proliferative effects in tumours with glycolytic phenotype, especially in case of IDH1 mutation (e.g. acute myeloid leukemias, gliomas, chondrosarcomas). Based on our new results, we suggest targeting mTOR activity depending on the metabolic and besides molecular genetic phenotype of tumours to increase the success of therapies.
Collapse
Affiliation(s)
- Zoltán Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Norbert Szoboszlai
- Laboratory of Environmental Chemistry and Bioanalytics, Department of Analytical Chemistry, Institute of Chemistry, Eötvös Loránd University, 1518, Budapest, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Noémi Nagy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Csilla Kriston
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Ildikó Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Sándor Paku
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary.,Tumor Progression Research Group of Joint Research Organization of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Olivér Ozohanics
- Hungarian Academy of Sciences, Research Centre for Natural Sciences, MS Proteomics Research Group, 1117, Budapest, Hungary
| | - László Drahos
- Hungarian Academy of Sciences, Research Centre for Natural Sciences, MS Proteomics Research Group, 1117, Budapest, Hungary
| | - András Jeney
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary. .,Tumor Progression Research Group of Joint Research Organization of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.
| |
Collapse
|
35
|
Zeng B, Liu L, Wang S, Dai Z. ILK regulates MSCs survival and angiogenesis partially through AKT and mTOR signaling pathways. Acta Histochem 2017; 119:400-406. [PMID: 28457660 DOI: 10.1016/j.acthis.2017.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/18/2017] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSCs) exert therapeutic effects on treating acute myocardial infarction (AMI). Angiogenesis in ischemic heart can promote the supply of oxygen and nutrients to both ischemic myocardium and transplanted stem cells. Focus is then given to the evolving strategies amied at angiogenesis. ILK has been reported to be an important factor regulating apoptosis and angiogenesis. This study examined the role and mechanism of ILK in MSCs survival and angiogenesis. In hypoxic condition, upregulation of ILK expression increased the phosphorylation of Akt and mTOR, resulting in markedly enchanced MSCs survival and VEGF expression; while significantly inhibited MSCs survival and VEGF expression was detected in MSCs with ILK kinase inactivation, which was associated with a reduction of phosphorylation of Akt and mTOR. In addition, it also caused an inhibitory effects of ILK on MSCs survival and VEGF expression, which was abolished by Akt or mTOR inhibitor. Furthermore, it was observed that ILK-overexpressed MSCs increased MSCs survival at 4days and angiogenesis at 3 weeks after transplantation into infracted myocardium as compared with GFP-MSCs group and ILK-SiRNA-MSCs group. This enhanced response was associated with attenuated left ventricular (LV) chamber dilation, reduced LV fibrosis, decreased infarct size and improved LV function. These findings reveal ILK play a pivotal role in regulating MSCs survival and VEGF expression partially through Akt and mTOR signaling pathway. In addition, transplantation of ILK-overexpressed MSCs into infracted myocardium resulted in reduced fibrosis, improved cardiac function and remodeling, which mainly medicated through increased MSCs survival and angiogenesis.
Collapse
Affiliation(s)
- Bin Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | - Lei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Shaofeng Wang
- Department of Internal Medicine, Jiangxia District Hospital of Traditional Chinese Medicine, Jiangxia, Hubei, PR China
| | - Zhiguo Dai
- The First People's Hospital of Cardiology, Jinmen, Hubei, PR China
| |
Collapse
|