1
|
Sun J, Sha M, Zhou J, Huang Y. Quercetin affects apoptosis and autophagy in pediatric acute myeloid leukaemia cells by inhibiting PI3K/AKT signaling pathway activation through regulation of miR-224-3p/PTEN axis. BMC Cancer 2025; 25:318. [PMID: 39984900 PMCID: PMC11843760 DOI: 10.1186/s12885-025-13709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
OBJECTIVE The aim of this study was to investigate the mechanism by which quercetin (Que) affects apoptosis and autophagy in pediatric acute myeloid leukaemia (AML) cells by inhibiting the activation of the PI3K/AKT signaling pathway through the regulation of the miR-224-3p/PTEN axis. METHODS Blood samples were collected from AML children and healthy volunteers. miR-224-3p and PTEN expression levels were measured. AML cells were pre-treated with Que. MiR-224-3p and PTEN expression levels in AML cells were altered via plasmid transfection. After intervention, PI3K/AKT phosphorylation, AML cell proliferation and apoptosis, concentrations of interleukin-1 β (IL-1β) and tumor necrosis factor-α (TNF-α) in AML cell culture supernatant, apoptosis-related genes Bax and Bcl-2, and autophagy markers LC3-I and LC3-II were tested. The targeting relationship between miR-224-3p and PTEN was identified. RESULTS MiR-224-3p expression was elevated in AML children, while PTEN was decreased. Que was available to accelerate AML cell apoptosis and restrain its autophagy. Que inhibited miR-224-3p expression and promoted PTEN expression. Upregulating miR-224-3p or downregulating PTEN weakened the effect of Que on AML cell apoptosis and autophagy. MiR-224-3p negatively modulated PTEN expression. Up-regulation of PTEN reversed the effects of up-regulation of miR-224-3p on apoptosis and autophagy in AML cells. In addition, Que inhibited PI3K/AKT signaling pathway activation, while up-regulation of miR-224-3p or down-regulation of PTEN could attenuate the inhibitory effect of Que on PI3K/AKT signaling pathway. Moreover, up-regulation of PTEN reversed the effect of up-regulation of miR-224-3p on the PI3K/AKT signaling pathway. CONCLUSION Que affects apoptosis and autophagy in pediatric AML cells by inhibiting PI3K/AKT signaling pathway activation through regulation of miR-224-3p/PTEN axis.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- PTEN Phosphohydrolase/metabolism
- PTEN Phosphohydrolase/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Apoptosis/drug effects
- Signal Transduction/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Autophagy/drug effects
- Quercetin/pharmacology
- Child
- Phosphatidylinositol 3-Kinases/metabolism
- Male
- Female
- Cell Proliferation/drug effects
- Cell Line, Tumor
- Child, Preschool
- Adolescent
- Gene Expression Regulation, Leukemic/drug effects
Collapse
Affiliation(s)
- Jing Sun
- Department of Pediatrics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, No.366, Taihu Road, Medical Hi-Tech Zone, Taizhou, Jiangsu, 225316, China.
| | - Min Sha
- Department of Central Laboratory, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, Taizhou, Jiangsu, 225316, China
| | - Jing Zhou
- Department of Oncology, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, Taizhou, Jiangsu, 225316, China
| | - Yun Huang
- Department of Pediatrics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, No.366, Taihu Road, Medical Hi-Tech Zone, Taizhou, Jiangsu, 225316, China
| |
Collapse
|
2
|
Ung J, Tan SF, Fox TE, Shaw JJP, Taori M, Horton BJ, Golla U, Sharma A, Szulc ZM, Wang HG, Chalfant CE, Cabot MC, Claxton DF, Loughran TP, Feith DJ. Acid Ceramidase Inhibitor LCL-805 Antagonizes Akt Signaling and Promotes Iron-Dependent Cell Death in Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:5866. [PMID: 38136410 PMCID: PMC10742122 DOI: 10.3390/cancers15245866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy requiring urgent treatment advancements. Ceramide is a cell-death-promoting signaling lipid that plays a central role in therapy-induced cell death. We previously determined that acid ceramidase (AC), a ceramide-depleting enzyme, is overexpressed in AML and promotes leukemic survival and drug resistance. The ceramidase inhibitor B-13 and next-generation lysosomal-localizing derivatives termed dimethylglycine (DMG)-B-13 prodrugs have been developed but remain untested in AML. Here, we report the in vitro anti-leukemic efficacy and mechanism of DMG-B-13 prodrug LCL-805 across AML cell lines and primary patient samples. LCL-805 inhibited AC enzymatic activity, increased total ceramides, and reduced sphingosine levels. A median EC50 value of 11.7 μM was achieved for LCL-805 in cell viability assays across 32 human AML cell lines. As a single agent tested across a panel of 71 primary AML patient samples, a median EC50 value of 15.8 μM was achieved. Exogenous ceramide supplementation with C6-ceramide nanoliposomes, which is entering phase I/II clinical trial for relapsed/refractory AML, significantly enhanced LCL-805 killing. Mechanistically, LCL-805 antagonized Akt signaling and led to iron-dependent cell death distinct from canonical ferroptosis. These findings elucidated key factors involved in LCL-805 cytotoxicity and demonstrated the potency of combining AC inhibition with exogenous ceramide.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Su-Fern Tan
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Todd E. Fox
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jeremy J. P. Shaw
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
| | - Maansi Taori
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
| | - Bethany J. Horton
- Department of Public Health Sciences, Division of Translational Research and Applied Statistics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Upendarrao Golla
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.F.C.)
| | - Arati Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Zdzislaw M. Szulc
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina College of Medicine, Charleston, SC 29425, USA;
| | - Hong-Gang Wang
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Charles E. Chalfant
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA 23249, USA
| | - Myles C. Cabot
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27858, USA;
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - David F. Claxton
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.F.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Thomas P. Loughran
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - David J. Feith
- Department of Medicine, Division of Hematology & Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (T.E.F.); (J.J.P.S.); (M.T.); (C.E.C.)
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
3
|
Combination Therapies Targeting Apoptosis in Paediatric AML: Understanding the Molecular Mechanisms of AML Treatments Using Phosphoproteomics. Int J Mol Sci 2023; 24:ijms24065717. [PMID: 36982791 PMCID: PMC10058112 DOI: 10.3390/ijms24065717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
Paediatric acute myeloid leukaemia (AML) continues to present treatment challenges, as no “standard approach” exists to treat those young patients reliably and safely. Combination therapies could become a viable treatment option for treating young patients with AML, allowing multiple pathways to be targeted. Our in silico analysis of AML patients highlighted “cell death and survival” as an aberrant, potentially targetable pathway in paediatric AML patients. Therefore, we aimed to identify novel combination therapies to target apoptosis. Our apoptotic drug screening resulted in the identification of one potential “novel” drug pairing, comprising the Bcl-2 inhibitor ABT-737 combined with the CDK inhibitor Purvalanol-A, as well as one triple combination of ABT-737 + AKT inhibitor + SU9516, which showed significant synergism in a series of paediatric AML cell lines. Using a phosphoproteomic approach to understand the apoptotic mechanism involved, proteins related to apoptotic cell death and cell survival were represented, in agreement with further results showing differentially expressed apoptotic proteins and their phosphorylated forms among combination treatments compared to single-agent treated cells such upregulation of BAX and its phosphorylated form (Thr167), dephosphorylation of BAD (Ser 112), and downregulation of MCL-1 and its phosphorylated form (Ser159/Thr 163). Total levels of Bcl-2 were decreased but correlated with increased levels of phosphorylated Bcl-2, which was consistent with our phosphoproteomic analysis predictions. Bcl-2 phosphorylation was regulated by extracellular-signal-regulated kinase (ERK) but not PP2A phosphatase. Although the mechanism linking to Bcl-2 phosphorylation remains to be determined, our findings provide first-hand insights on potential novel combination treatments for AML.
Collapse
|
4
|
Castelletti F, Consonni G. Bayesian graphical modeling for heterogeneous causal effects. Stat Med 2023; 42:15-32. [PMID: 36317356 DOI: 10.1002/sim.9599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 09/08/2022] [Accepted: 10/15/2022] [Indexed: 12/24/2022]
Abstract
There is a growing interest in current medical research to develop personalized treatments using a molecular-based approach. The broad goal is to implement a more precise and targeted decision-making process, relative to traditional treatments based primarily on clinical diagnoses. Specifically, we consider patients affected by Acute Myeloid Leukemia (AML), an hematological cancer characterized by uncontrolled proliferation of hematopoietic stem cells in the bone marrow. Because AML responds poorly to chemotherapeutic treatments, the development of targeted therapies is essential to improve patients' prospects. In particular, the dataset we analyze contains the levels of proteins involved in cell cycle regulation and linked to the progression of the disease. We evaluate treatment effects within a causal framework represented by a Directed Acyclic Graph (DAG) model, whose vertices are the protein levels in the network. A major obstacle in implementing the above program is represented by individual heterogeneity. We address this issue through a Dirichlet Process (DP) mixture of Gaussian DAG-models where both the graphical structure as well as the allied model parameters are regarded as uncertain. Our procedure determines a clustering structure of the units reflecting the underlying heterogeneity, and produces subject-specific estimates of causal effects based on Bayesian Model Averaging (BMA). With reference to the AML dataset, we identify different effects of protein regulation among individuals; moreover, our method clusters patients into groups that exhibit only mild similarities with traditional categories based on morphological features.
Collapse
Affiliation(s)
- Federico Castelletti
- Department of Statistical Sciences, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Guido Consonni
- Department of Statistical Sciences, Università Cattolica del Sacro Cuore, Milan, Italy
| |
Collapse
|
5
|
Centio A, Estruch M, Reckzeh K, Sanjiv K, Vittori C, Engelhard S, Warpman Berglund U, Helleday T, Theilgaard-Mönch K. Inhibition of Oxidized Nucleotide Sanitation By TH1579 and Conventional Chemotherapy Cooperatively Enhance Oxidative DNA Damage and Survival in AML. Mol Cancer Ther 2022; 21:703-714. [PMID: 35247918 DOI: 10.1158/1535-7163.mct-21-0185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
Currently, the majority of patients with acute myeloid leukemia (AML) still die of their disease due to primary resistance or relapse toward conventional reactive oxygen species (ROS)- and DNA damage-inducing chemotherapy regimens. Herein, we explored the therapeutic potential to enhance chemotherapy response in AML, by targeting the ROS scavenger enzyme MutT homolog 1 (MTH1, NUDT1), which protects cellular integrity through prevention of fatal chemotherapy-induced oxidative DNA damage. We demonstrate that MTH1 is a potential druggable target expressed by the majority of patients with AML and the inv(16)/KITD816Y AML mouse model mimicking the genetics of patients with AML exhibiting poor response to standard chemotherapy (i.e., anthracycline & cytarabine). Strikingly, combinatorial treatment of inv(16)/KITD816Y AML cells with the MTH1 inhibitor TH1579 and ROS- and DNA damage-inducing standard chemotherapy induced growth arrest and incorporated oxidized nucleotides into DNA leading to significantly increased DNA damage. Consistently, TH1579 and chemotherapy synergistically inhibited growth of clonogenic inv(16)/KITD816Y AML cells without substantially inhibiting normal clonogenic bone marrow cells. In addition, combinatorial treatment of inv(16)/KITD816Y AML mice with TH1579 and chemotherapy significantly reduced AML burden and prolonged survival compared with untreated or single treated mice. In conclusion, our study provides a rationale for future clinical studies combining standard AML chemotherapy with TH1579 to boost standard chemotherapy response in patients with AML. Moreover, other cancer entities treated with ROS- and DNA damage-inducing chemo- or radiotherapies might benefit therapeutically from complementary treatment with TH1579.
Collapse
Affiliation(s)
- Anders Centio
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Montserrat Estruch
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Kristian Reckzeh
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, Centre for Stem Cell Research and Developmental Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kumar Sanjiv
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Camilla Vittori
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Sophia Engelhard
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Ulrika Warpman Berglund
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Kim Theilgaard-Mönch
- The Finsen Laboratory, Centre for Cancer and Organ Diseases, Rigshospitalet/National University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biotech Research and Innovation Center, Faculty of Health and Medical Sciences (BRIC), University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Stem Cell Biology, Centre for Stem Cell Research and Developmental Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Hematology, Rigshospitalet/National University Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Pfeiffer A, Franciosa G, Locard-Paulet M, Piga I, Reckzeh K, Vemulapalli V, Blacklow SC, Theilgaard-Mönch K, Jensen LJ, Olsen JV. Phosphorylation of SHP2 at Tyr62 enables acquired resistance to SHP2 allosteric inhibitors in FLT3-ITD-driven AML. Cancer Res 2022; 82:2141-2155. [PMID: 35311954 DOI: 10.1158/0008-5472.can-21-0548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 12/17/2021] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
The protein tyrosine phosphatase SHP2 is crucial for oncogenic transformation of acute myeloid leukemia (AML) cells expressing mutated receptor tyrosine kinases (RTK). SHP2 is required for full RAS-ERK activation to promote cell proliferation and survival programs. Allosteric SHP2 inhibitors act by stabilizing SHP2 in its auto-inhibited conformation and are currently being tested in clinical trials for tumors with overactivation of the RAS/ERK pathway, alone and in various drug combinations. In this study, we established cells with acquired resistance to the allosteric SHP2 inhibitor SHP099 from two FLT3-ITD-positive AML cell lines. Label-free and isobaric labeling quantitative mass spectrometry-based phosphoproteomics of these resistant models demonstrated that AML cells can restore phosphorylated ERK (pERK) in the presence of SHP099, thus developing adaptive resistance. Mechanistically, SHP2 inhibition induced tyrosine phosphorylation and feedback-driven activation of the FLT3 receptor, which in turn phosphorylated SHP2 on tyrosine 62. This phosphorylation stabilized SHP2 in its open conformation, preventing SHP099 binding and conferring resistance. Combinatorial inhibition of SHP2 and MEK or FLT3 prevented pERK rebound and resistant cell growth. The same mechanism was observed in a FLT3-mutated B-ALL cell line and in the inv(16)/KitD816Y AML mouse model, but allosteric inhibition of Shp2 did not impair the clonogenic ability of normal bone marrow progenitors. Together, these results support the future use of SHP2 inhibitor combinations for clinical applications.
Collapse
Affiliation(s)
| | | | | | - Ilaria Piga
- Istituto Oncologico Veneto IOV - IRCCS, Italy
| | | | | | | | | | | | | |
Collapse
|
7
|
Zhang R, Ozgen S, Luo H, Krigman J, Zhao Y, Xin G, Sun N. The Mitochondrial Deubiquitinase USP30 Regulates AKT/mTOR Signaling. Front Pharmacol 2022; 13:816551. [PMID: 35250566 PMCID: PMC8891576 DOI: 10.3389/fphar.2022.816551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Mitophagy is an intracellular mechanism to maintain mitochondrial health by removing dysfunctional mitochondria. The E3 ligase Parkin ubiquitinates the membrane proteins on targeted mitochondria to initiate mitophagy, whereas USP30 antagonizes Parkin-dependent mitophagy by removing ubiquitin from Parkin substrates. The AKT/mTOR signaling is a master regulator of cell proliferation, differentiation, apoptosis, and autophagy. Although mounting evidence suggests that perturbations in the AKT/mTOR signaling pathway may contribute to mitophagy regulation, the specific mechanisms between Parkin/USP30 and AKT/mTOR signaling have not been elucidated. In this study, we employ a set of genetic reagents to investigate the role of Parkin and USP30 in regulating the AKT/mTOR signaling during mitophagy. We demonstrated that, in the setting of mitochondrial stress, the AKT/mTOR signaling is regulated, at least in part, by the activity of Parkin and USP30. Parkin inhibits AKT/mTOR signaling following an in vitro mitochondrial stress, thereby promoting apoptosis. However, USP30 overexpression antagonizes the activity of Parkin to sustain AKT/mTOR activity and inhibit apoptosis. These findings provide new insights into Parkin and USP30’s role in apoptosis and suggest that inhibiting USP30 might provide a specific strategy to synergize with AKT/mTOR inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Ruohan Zhang
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, United States
| | - Serra Ozgen
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Hongke Luo
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Judith Krigman
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Yutong Zhao
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Gang Xin
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Nuo Sun
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- *Correspondence: Nuo Sun,
| |
Collapse
|
8
|
Guo Y, Wang L, Yang H, Ding N. Knockdown long non-coding RNA HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation. Bioengineered 2022; 13:884-893. [PMID: 34969363 PMCID: PMC8805942 DOI: 10.1080/21655979.2021.2014386] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Recently, long noncoding RNAs (lncRNAs) have been revealed to participate in cancer therapy. Especial in tumor radiotherapy, lncRNAs usually could enhance or restrict the radiosensitivity in different ways. LncRNA HCP5 is highly expressed in esophageal cancer and influenced the malignant behaviors of esophageal cancer cells. However, this study dedicates to clarify if lncRNA HCP5 affects the radiosensitivity of esophageal carcinoma. The expression levels of HCP5 in esophageal cancer and adjacent noncancerous tissue were first analyzed on the TCGA database and then detected by qRT-PCR. The related functional experiments were used to investigate whether the radiosensitivity of esophageal squamous cell carcinoma was affected by the inhibition of HCP5. The expression results showed HCP5 is upregulated in esophageal cancers compared to the normal tissues. Meanwhile, knockdown HCP5 further suppressed the proliferation and promoted the apoptosis of esophageal cancer cells treated with a 2 Gy dose of radiotherapy. Moreover, we uncovered that knockdown HCP5 eliminated radiotherapy resistance by modulating the miR-216a-3p/PDK1 axis to inhibit the AKT activation. Finally, rescue experiments pointed that lowering the miR-216a-3p expression weakened the inhibition effect of knockdown HCP5 on cells treated with radiotherapy. To summary, our results indicate that HCP5 is involved in esophageal carcinoma radiotherapy and knockdown HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation.
Collapse
Affiliation(s)
- Yue Guo
- Hematology Department, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Lan Wang
- Department of Anesthesiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Hui Yang
- Department of Anesthesiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Nannan Ding
- Department of Pharmacy, Xiangyang Central Hospital, Affiliated of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
9
|
Zeng YJ, Wu M, Zhang H, Wu XP, Zhou L, Wan N, Wu ZH. Effects of Qinghuang Powder on Acute Myeloid Leukemia Based on Network Pharmacology, Molecular Docking, and In Vitro Experiments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6195174. [PMID: 34992668 PMCID: PMC8727110 DOI: 10.1155/2021/6195174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/09/2023]
Abstract
Qinghuang powder (QHP) is a traditional Chinese herbal medicine. This is a unique formula that is frequently used to treat malignant hematological diseases such as acute myeloid leukemia (AML) in modern clinical practice. An approach of network pharmacology and experimental validation were applied to investigate the pharmacological mechanisms of QHP in AML treatment. First, public databases for target genes known to be associated with AML are searched and compared to the target genes of the active compounds in QHP. Second, AML-associated genes and QHP target genes are compared to identify overlapping enriched genes, and these were used to predict selected target genes that may be implicated in the effects of QHP on AML. Additionally, we conducted functional enrichment analyses, such as gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The significantly enriched pathway associated with potential target proteins was the PI3K-Akt signaling pathway, suggesting that these potential target proteins and pathways may mediate the beneficial biological effects of QHP on AML. All these following genes were found to occur in the compounds-target-pathway networks: AKT1, MAPK1, MAPK3, PIK3CG, CASP3, CASP9, TNF, TGFB1, MAPK8, and TP53. Then, based on the molecular docking studies, it was suggested that the active compound isovitexin can fit into the binding pockets of the top candidate QHP-AML target proteins (PIK3CG). Subsequently, based on the prediction by network pharmacology analysis, both in vitro AML cells and western blot experiments were performed to validate the curative role of QHP. QHP exerted its antitumor activity on AML in vitro, as it inhibits cells proliferation, reduced the expression of Bcl-2 protein, and downregulated the PI3K-Akt signaling pathway. In conclusion, these results revealed that QHP could treat AML via a "multicomponent, multitarget, multipathway" regulatory network. Furthermore, our study also demonstrated that the combination of network pharmacology with the experimental study is effective in discovering and identifying QHP in the treatment of AML and its underlying pharmacological mechanisms.
Collapse
Affiliation(s)
- Ying-jian Zeng
- Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Min Wu
- Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Huan Zhang
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Xin-ping Wu
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Lu Zhou
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Na Wan
- Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Zhen-hui Wu
- Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
- The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
10
|
Jaffredo T, Balduini A, Bigas A, Bernardi R, Bonnet D, Canque B, Charbord P, Cumano A, Delwel R, Durand C, Fibbe W, Forrester L, de Franceschi L, Ghevaert C, Gjertsen B, Gottgens B, Graf T, Heidenreich O, Hermine O, Higgs D, Kleanthous M, Klump H, Kouskoff V, Krause D, Lacaud G, Celso CL, Martens JH, Méndez-Ferrer S, Menendez P, Oostendorp R, Philipsen S, Porse B, Raaijmakers M, Robin C, Stunnenberg H, Theilgaard-Mönch K, Touw I, Vainchenker W, Corrons JLV, Yvernogeau L, Schuringa JJ. The EHA Research Roadmap: Normal Hematopoiesis. Hemasphere 2021; 5:e669. [PMID: 34853826 PMCID: PMC8615310 DOI: 10.1097/hs9.0000000000000669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Thierry Jaffredo
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | | | - Anna Bigas
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Bernardi
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Bruno Canque
- INSERM U976, Universite de Paris, Ecole Pratique des Hautes Etudes/PSL Research University, Institut de Recherche Saint Louis, France
| | - Pierre Charbord
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Anna Cumano
- Unité Lymphopoïèse, Département d’Immunologie, INSERM U1223, Institut Pasteur, Cellule Pasteur, Université de Paris, France
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Charles Durand
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Willem Fibbe
- Leiden University Medical Center, The Netherlands
| | - Lesley Forrester
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Scotland
| | | | | | - Bjørn Gjertsen
- Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Berthold Gottgens
- Wellcome - MRC Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, United Kingdom
| | - Thomas Graf
- Center for Genomic Regulation, Barcelona Institute for Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain
| | - Olaf Heidenreich
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Olivier Hermine
- Department of Hematology and Laboratory of Physiopathology and Treatment of Blood Disorders, Hôpital Necker, Imagine institute, University of Paris, France
| | - Douglas Higgs
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | | | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | | | - Daniela Krause
- Goethe University Frankfurt and Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - George Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | | | - Joost H.A. Martens
- Department of Molecular Biology, RIMLS, Radboud University, Nijmegen, The Netherlands
| | | | - Pablo Menendez
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-RETAV, Instituto de Salud Carlos III, Madrid, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - Robert Oostendorp
- Department of Internal Medicine III, Technical University of Munich, School of Medicine, Germany
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Bo Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Marc Raaijmakers
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW and University Medical Center Utrecht, The Netherlands
- Regenerative medicine center, University Medical Center Utrecht, The Netherlands
| | - Henk Stunnenberg
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Kim Theilgaard-Mönch
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
- Department of Hematology, Rigshospitalet/National University Hospital, University of Copenhagen, Denmark
| | - Ivo Touw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Joan-Lluis Vives Corrons
- Red Blood Cell and Hematopoietic Disorders Research Unit, Institute for Leukaemia Research Josep Carreras, Badalona, Barcelona
| | - Laurent Yvernogeau
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, The Netherlands
| |
Collapse
|