1
|
Yang X, Wang Y, Feng Y, Wang F, Gao L, Wang Y. Trimethylamine oxide promotes myocardial fibrosis through activating JAK2-STAT3 pathway. Biochem Biophys Res Commun 2025; 750:151390. [PMID: 39889626 DOI: 10.1016/j.bbrc.2025.151390] [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: 01/14/2025] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Myocardial infarction is often accompanied by symptoms of myocardial fibrosis, causing myocardial systolic and diastolic dysfunction. High levels of TMAO are considered to be strongly connected to cardiovascular diseases occurrence. The objective of this investigation was to determine whether formal TMAO affects the development of cardiac muscle fibers. The left anterior descending (LAD) coronary artery was ligated to construct myocardial infarction mouse model, while the control group mouse was only operated vessel separation. According to whether or not TMAO intervention is given, mice were divided into control, TMAO, myocardial infarction model(model), and myocardial infarction model + TMAO(model + TMAO). TMAO and model + TMAO group received 0.24 % TMAO in drinking water for one month. After modeling, echocardiography assessed cardiac function, and cardiac tissues were collected for analysis. Masson staining observed collagen deposition, while immunohistochemistry was conducted to identify fibronectin, collagen III, collagen I, and JAK/STAT pathway related proteins levels. In vitro, mouse cardiac fibroblasts were cultured and treated with varying TMAO concentrations (0, 300, 600, 900 μM), and Western blot was carried out to analyse changes in fibronectin, collagen III, and collagen I. Subsequently, the JAK2/STAT3 pathway inhibitor AG490 was utilized to treat cells and further examine the changes of fibronectin and collagen. Thein vivodata showed TMAO significantly increases fibrosis and upregulates fibronectin, collagen III, and collagen I.In vitroexperiments indicated that TMAO promotes fibronectin, collagen III, and collagen I levels through regulating the JAK2/STAT3 pathway, ultimately accelerating fibrosis.
Collapse
Affiliation(s)
- Xiaoyun Yang
- The Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yi Wang
- The Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yujin Feng
- The Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Feng Wang
- The Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lijuan Gao
- The Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yueheng Wang
- The Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
2
|
Jian J, Wang N, Hao H, Yuan C, Liu Q, Ji C, Lu F. SLED1 Promoting Cell Proliferation and Inhibiting Apoptosis in Acute Myeloid Leukemia: a Study. Appl Biochem Biotechnol 2023; 195:6633-6652. [PMID: 36897493 DOI: 10.1007/s12010-023-04421-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/11/2023]
Abstract
In this study, we aimed to explore long non-coding RNA (lncRNA) sustained low-efficiency dialysis (SLED1) correlated with Bcl-2 apoptosis pathway in acute myeloid leukemia (AML). This study further aimed to determine its role in the regulation of AML progression and its action as a potential biomarker for better prognosis. AML microarray profiles GSE97485 and probe annotation from the Gene Expression Omnibus (GEO) database from the National Center for Biotechnology Information (NCBI) were detected using the GEO2R tool ( http://www.ncbi.nlm.nih.gov/geo/geo2r/ ). The expression of AML was downloaded from the TCGA database ( http://cancergenome.nih.gov/ ). The statistical analysis of the database was processed with R software. Bioinformatic analysis found that lncRNA SLED1 is highly expressed in AML patients and is associated with poor prognosis. We found that the increased SLED1 expression levels in AML were significantly correlated with FAB classification, human race, and age. Our study has shown that upregulation of SLED1 promoted AML cell proliferation and inhibited cell apoptosis in vitro; RNA sequencing showed increased expression of BCL-2 and indicated that SLED1 might promote the development of AML by regulating BCL-2. Our results showed that SLED1 could promote the proliferation and inhibit the apoptosis of AML cells. SLED1 might promote the development of AML by regulating BCL-2, but the mechanism involved in the progression of AML is unclear. SLED1 plays an important role in AML progression, may be applied as a rapid and economical AML prognostic indicator to predict the survival of AML patients, and help guide experiments for potential clinical drag targets.
Collapse
Affiliation(s)
- Jimo Jian
- Department of Hematology, Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Nana Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Hongyuan Hao
- Department of Hematology, Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
| | - Chenglu Yuan
- Department of Hematology, Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
| | - Qian Liu
- Department of Pain, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fei Lu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
3
|
Yang G, Tong Y, Wang X, Zhao C, Ba Z, Ahelijiang R, Liu X, Gao W, Zhao Y, Gu Y, Yang J, Xu Y. Guizhi Fuling capsule relieves memory deficits by inhibition of microglial neuroinflammation through blocking JAK2/STAT3 pathway in presenilin1/2 conditional double knockout mice. Front Immunol 2023; 14:1185570. [PMID: 37465679 PMCID: PMC10350565 DOI: 10.3389/fimmu.2023.1185570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic neuroinflammation has been regarded as an important part of the pathological initiation of Alzheimer's disease (AD), which is associated with the regulation of microglial activation. Preventing microglial activation to inhibit neuroinflammation may become a potential target for the treatment of neurodegenerative diseases. Guizhi Fuling capsule (GZFL) has a strong repression on inflammatory responses. Here, the presenilin1/2 conditional double knockout (PS cDKO) mice, a well-established mouse model of AD, were divided into: WT mice (WT), WT mice+GZFL (WT+GZFL), PS cDKO mice (cDKO), and PS cDKO mice+GZFL (cDKO+GZFL). Mice in the WT+GZFL and cDKO+GZFL group were fed standard chow containing 2000 ppm GZFL for 90 days. After 60 days of GZFL treatment, mice were given to behavioral tests for 30 days in order to explore the effects of GZFL on cognitive and motor function. Then, mice were sacrificed for examining the effects of GZFL on inflammation. Furthermore, primary microglia were obtained from neonatal Sprague-Dawley rats and pretreated with or without GZFL (50 μg/ml) for 1 h in the absence or presence of lipopolysaccharide (LPS) (100 ng/ml) stimulation to speculate whether the underlying mechanism of GZFL's anti-inflammatory potential was closely associated with Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Our findings indicated that GZFL has the ability to alleviate memory deficits in PS cDKO mice, which attributes to the improvement of neuroinflammation by inhibiting microglial activation and the levels of pro-inflammatory mediators. In addition, GZFL could inverse the tau hyperphosphorylation and the lessened expression of synaptic proteins in hippocampus of PS cDKO mice. Furthermore, GZFL prevented LPS-induced neuroinflammatory responses in primary microglia by decreasing the levels of pro-inflammatory mediators. It is noteworthy that therapeutic effects of GZFL on memory impairment are depended on the inhibition of neuroinflammatory responses by the blockage of JAK2/STAT3 signaling pathway. Taken together, GZFL may be an effective compound Chinese medicine for the improvement and postponement of neurodegenerative progression in AD.
Collapse
Affiliation(s)
- Guang Yang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuting Tong
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xingyu Wang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongtao Ba
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Reaila Ahelijiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinjuan Liu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Waimao Gao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yining Gu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Yangzhi Rehabilitation Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianmei Yang
- Department of Traditional Chinese Medicine, Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Palamarchuk AI, Kovalenko EI, Streltsova MA. Multiple Actions of Telomerase Reverse Transcriptase in Cell Death Regulation. Biomedicines 2023; 11:biomedicines11041091. [PMID: 37189709 DOI: 10.3390/biomedicines11041091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Telomerase reverse transcriptase (TERT), a core part of telomerase, has been known for a long time only for its telomere lengthening function by reverse transcription of RNA template. Currently, TERT is considered as an intriguing link between multiple signaling pathways. The diverse intracellular localization of TERT corresponds to a wide range of functional activities. In addition to the canonical function of protecting chromosome ends, TERT by itself or as a part of the telomerase complex participates in cell stress responses, gene regulation and mitochondria functioning. Upregulation of TERT expression and increased telomerase activity in cancer and somatic cells relate to improved survival and persistence of such cells. In this review, we summarize the data for a comprehensive understanding of the role of TERT in cell death regulation, with a focus on the interaction of TERT with signaling pathways involved in cell survival and stress response.
Collapse
Affiliation(s)
- Anastasia I. Palamarchuk
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Elena I. Kovalenko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria A. Streltsova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
5
|
Ivanenko KA, Prassolov VS, Khabusheva ER. Transcription Factor Sp1 in the Expression of Genes Encoding Components of Mapk, JAK/STAT, and PI3K/Akt Signaling Pathways. Mol Biol 2022. [DOI: 10.1134/s0026893322050089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
6
|
Khokhlatchev AV, Sharma A, Deering TG, Shaw JJP, Costa‐Pinheiro P, Golla U, Annageldiyev C, Cabot MC, Conaway MR, Tan S, Ung J, Feith DJ, Loughran TP, Claxton DF, Fox TE, Kester M. Ceramide nanoliposomes augment the efficacy of venetoclax and cytarabine in models of acute myeloid leukemia. FASEB J 2022; 36:e22514. [PMID: 36106439 PMCID: PMC9544744 DOI: 10.1096/fj.202200765r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 12/12/2022]
Abstract
Despite several new therapeutic options for acute myeloid leukemia (AML), disease relapse remains a significant challenge. We have previously demonstrated that augmenting ceramides can counter various drug-resistance mechanisms, leading to enhanced cell death in cancer cells and extended survival in animal models. Using a nanoscale delivery system for ceramide (ceramide nanoliposomes, CNL), we investigated the effect of CNL within a standard of care venetoclax/cytarabine (Ara-C) regimen. We demonstrate that CNL augmented the efficacy of venetoclax/cytarabine in in vitro, ex vivo, and in vivo models of AML. CNL treatment induced non-apoptotic cytotoxicity, and augmented cell death induced by Ara-C and venetoclax. Mechanistically, CNL reduced both venetoclax (Mcl-1) and cytarabine (Chk1) drug-resistant signaling pathways. Moreover, venetoclax and Ara-C augmented the generation of endogenous pro-death ceramide species, which was intensified with CNL. Taken together, CNL has the potential to be utilized as an adjuvant therapy to improve outcomes, potentially extending survival, in patients with AML.
Collapse
Affiliation(s)
| | - Arati Sharma
- Division of Hematology and Oncology, Department of MedicinePenn State University College of MedicineHersheyPennsylvaniaUSA
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Penn State Cancer InstitutePennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Tye G. Deering
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Jeremy J. P. Shaw
- Department of Experimental PathologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pedro Costa‐Pinheiro
- Department of Experimental PathologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Upendarrao Golla
- Division of Hematology and Oncology, Department of MedicinePenn State University College of MedicineHersheyPennsylvaniaUSA
- Penn State Cancer InstitutePennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Charyguly Annageldiyev
- Division of Hematology and Oncology, Department of MedicinePenn State University College of MedicineHersheyPennsylvaniaUSA
- Penn State Cancer InstitutePennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Myles C. Cabot
- Department of Biochemistry and Molecular Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Mark R. Conaway
- University of Virginia School of MedicinePublic Health SciencesCharlottesvilleVirginiaUSA
| | - Su‐Fern Tan
- Division of Hematology and Oncology, Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- University of Virginia Cancer CenterCharlottesvilleVirginiaUSA
| | - Johnson Ung
- Division of Hematology and Oncology, Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Department of Microbiology, Immunology and Cancer BiologyUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - David J. Feith
- Division of Hematology and Oncology, Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- University of Virginia Cancer CenterCharlottesvilleVirginiaUSA
| | - Thomas P. Loughran
- Division of Hematology and Oncology, Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- University of Virginia Cancer CenterCharlottesvilleVirginiaUSA
| | - David F. Claxton
- Division of Hematology and Oncology, Department of MedicinePenn State University College of MedicineHersheyPennsylvaniaUSA
- Penn State Cancer InstitutePennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Todd E. Fox
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Mark Kester
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- University of Virginia Cancer CenterCharlottesvilleVirginiaUSA
- NanoSTAR InstituteCharlottesvilleVirginiaUSA
| |
Collapse
|
7
|
Chen Z, Zhou H, Huang X, Wang S, Ouyang X, Wang Y, Cao Q, Yang L, Tao Y, Lai H. Pirfenidone attenuates cardiac hypertrophy against isoproterenol by inhibiting activation of the janus tyrosine kinase-2/signal transducer and activator of transcription 3 (JAK-2/STAT3) signaling pathway. Bioengineered 2022; 13:12772-12782. [PMID: 35609321 PMCID: PMC9276057 DOI: 10.1080/21655979.2022.2073145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiovascular risk factors have attracted increasing attention in recent years with the acceleration of population aging, amongst which cardiac hypertrophy is the initiating link to heart failure. Pirfenidone is a promising agent for the treatment of idiopathic pulmonary fibrosis and has recently proven to exert inhibitory effects on the inflammatory response. This study proposes to explore the potential pharmacological action of pirfenidone in treating cardiac hypertrophy in a rodent model. Four groups of mice were used in the present study: the control, ISO (5 mg/kg/day) for 7 days, pirfenidone (200 mg/kg/day) for 14 days, and spironolactone (SPI) (200 mg/kg/day) for 14 days groups. Increased heart weight index, left ventricle (LV) weight index, LV wall thickness, declined LV volume, and elevated serum levels of CK-MB, AST, and LDH were observed in ISO-challenged mice, all of which were dramatically reversed by the administration of pirfenidone or SPI. Furthermore, an elevated cross-sectional area of cardiomyocytes in the wheat germ agglutinin (WGA) staining of heart cross-sections, upregulated atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), β-Myosin Heavy Chain (β-MHC), and excessively released tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6) in cardiac tissues were observed in the ISO group but greatly alleviated by pirfenidone or SPI. Lastly, the promoted expression levels of p-JAK-2/JAK-2 and p-STAT3/STAT-3 in the cardiac tissues of ISO-challenged mice were significantly repressed by pirfenidone or SPI. Collectively, our data reveals a therapeutic property of pirfenidone on ISO-induced cardiac hypertrophy in mice.
Collapse
Affiliation(s)
- Zhenhuan Chen
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Haiwen Zhou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Xiantao Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Shaoyun Wang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Xiaochao Ouyang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yunxia Wang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qianqiang Cao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Liu Yang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yu Tao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Hengli Lai
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Pasca S, Chifotides HT, Verstovsek S, Bose P. Mutational landscape of blast phase myeloproliferative neoplasms (MPN-BP) and antecedent MPN. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:83-124. [PMID: 35153007 DOI: 10.1016/bs.ircmb.2021.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Myeloproliferative neoplasms (MPN) have an inherent tendency to evolve to the blast phase (BP), characterized by ≥20% myeloblasts in the blood or bone marrow. MPN-BP portends a dismal prognosis and currently, effective treatment modalities are scarce, except for allogeneic hematopoietic stem cell transplantation (allo-HSCT) in selected patients, particularly those who achieve complete/partial remission. The mutational landscape of MPN-BP differs from de novo acute myeloid leukemia (AML) in several key aspects, such as significantly lower frequencies of FLT3 and DNMT3A mutations, and higher incidence of IDH1/2 and TP53 in MPN-BP. Herein, we comprehensively review the impact of the three signaling driver mutations (JAK2 V617F, CALR exon 9 indels, MPL W515K/L) that constitutively activate the JAK/STAT pathway, and of the other somatic non-driver mutations (epigenetic, mRNA splicing, transcriptional regulators, and mutations in signal transduction genes) that cooperatively or independently promote MPN progression and leukemic transformation. The MPN subtype, harboring two or more high-molecular risk (HMR) mutations (epigenetic regulators and mRNA splicing factors) and "triple-negative" PMF are among the critical factors that increase risk of leukemic transformation and shorten survival. Primary myelofibrosis (PMF) is the most aggressive MPN; and polycythemia vera (PV) and essential thrombocythemia (ET) are relatively indolent subtypes. In PV and ET, mutations in splicing factor genes are associated with progression to myelofibrosis (MF), and in ET, TP53 mutations predict risk for leukemic transformation. The advent of targeted next-generation sequencing and improved prognostic scoring systems for PMF inform decisions regarding allo-HSCT. The emergence of treatments targeting mutant enzymes (e.g., IDH1/2 inhibitors) or epigenetic pathways (BET and LSD1 inhibitors) along with new insights into the mechanisms of leukemogenesis will hopefully lead the way to superior management strategies and outcomes of MPN-BP patients.
Collapse
Affiliation(s)
- Sergiu Pasca
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Helen T Chifotides
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Srdan Verstovsek
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Prithviraj Bose
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
9
|
Zhou Q, Lin L, Li H, Wang H, Jiang S, Huang P, Lin Q, Chen X, Deng Y. Melatonin Reduces Neuroinflammation and Improves Axonal Hypomyelination by Modulating M1/M2 Microglia Polarization via JAK2-STAT3-Telomerase Pathway in Postnatal Rats Exposed to Lipopolysaccharide. Mol Neurobiol 2021; 58:6552-6576. [PMID: 34585328 PMCID: PMC8639545 DOI: 10.1007/s12035-021-02568-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/12/2021] [Indexed: 02/05/2023]
Abstract
Microglia activation and associated inflammation are implicated in the periventricular white matter damage (PWMD) in septic postnatal rats. This study investigated whether melatonin would mitigate inflammation and alleviate the axonal hypomyelination in the corpus callosum in septic postnatal rats. We further explored if this might be related to the modulation of microglial polarization from M1 phenotype to M2 through the JAK2/STAT3/telomerase pathway. We reported here that indeed melatonin not only can it reduce the neurobehavioral disturbances in LPS-injected rats, but it can also dampen microglia-mediated inflammation. Thus, in LPS + melatonin group, the expression of proinflammatory mediators in M1 phenotype microglia was downregulated. As opposed to this, M2 microglia were increased which was accompanied by upregulated expression of anti-inflammatory mediators along with telomerase reverse transcriptase or melatonin receptor 1(MT1). In parallel to this was decreased NG2 expression but increased expression of myelin and neurofilament proteins. Melatonin can improve hypomyelination which was confirmed by electron microscopy. In vitro in primary microglia stimulated by LPS, melatonin decreased the expression of proinflammatory mediators significantly; but it increased the expression of anti-inflammatory mediators. Additionally, the expression levels of p-JAK2 and p-STAT3 were significantly elevated in microglia after melatonin treatment. Remarkably, the effect of melatonin on LPS-treated microglia was blocked by melatonin receptor, JAK2, STAT3 and telomerase reverse transcriptase inhibitors, respectively. Taken together, it is concluded that melatonin can attenuate PWMD through shifting M1 microglia towards M2 via MT1/JAK2/STAT3/telomerase pathway. The results suggest a new therapeutic strategy whereby melatonin may be adopted to convert microglial polarization from M1 to M2 phenotype that would ultimately contribute to the attenuation of PWMD.
Collapse
Affiliation(s)
- Qiuping Zhou
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Lanfen Lin
- Department of Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Haiyan Li
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Huifang Wang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Shuqi Jiang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Peixian Huang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qiongyu Lin
- Department of Critical Care Medicine, Jieyang People's Hospital, Jieyang, 522000, Guangdong, China
| | - Xuan Chen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Shantou University Medical College (FCS), Shantou, 515063, China
| | - Yiyu Deng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
10
|
Bakhshayesh M, Gohari LH, Barati M, Safa M. Combination therapy using TGF-β1 and STI-571 can induce apoptosis in BCR-ABL oncogene-expressing cells. Biomol Concepts 2021; 12:144-155. [PMID: 34700368 DOI: 10.1515/bmc-2021-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/24/2021] [Indexed: 11/15/2022] Open
Abstract
The BCR-ABL oncogene is a tyrosine kinase gene that is over-expressed in CML. It inhibits the TGF-β1 signaling pathway. Due to resistance of cells to the tyrosine kinase inhibitor, STI-571, the combined effect of STI-571 and TGF-β1 on K562 cells was studied in the present research. Results revealed that the TGF-β1 cell signaling pathway, which is activated in K562 cells treated with TGF-β1, activates collective cell signaling pathways involved in survival and apoptosis. It is noteworthy that treating K562 cells with STI-571 triggered apoptotic pathways, accompanied by a reduction in proteins such as Bcl-xL, Bcl-2, p-AKT, p-Stat5, p-FOXO3, and Mcl-1 and an increase in the pro-apoptotic proteins PARP cleavage, and p27, leading to an increase in sub-G1 phase-arrested and Annexin-positive cells. Interestingly, the proliferation behavior of TGF-β1-induced cells was changed with the combination therapy, and STI-571-induced apoptosis was also prompted by this combination. Thus, combination treatment appears to promote sub-G1 cell cycle arrest compared to individually treated cells. Furthermore, it strongly triggered apoptotic signaling. In conclusion, TGF-β1 did not negatively impact the effect of STI-571, based on positive annexin cells, and AKT protein phosphorylation remains effective in apoptosis.
Collapse
Affiliation(s)
- Masoome Bakhshayesh
- Genetics department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ladan Hosseini Gohari
- Cellular & Molecular Research Center, Medical Laboratory Science Department, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Safa
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Watkinson F, Nayar SK, Rani A, Sakellariou CA, Elhage O, Papaevangelou E, Dasgupta P, Galustian C. IL-15 Upregulates Telomerase Expression and Potently Increases Proliferative Capacity of NK, NKT-Like, and CD8 T Cells. Front Immunol 2021; 11:594620. [PMID: 33537030 PMCID: PMC7848219 DOI: 10.3389/fimmu.2020.594620] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Interleukin-15 (IL-15) is a cytokine that has been shown to expand CD8 T cell and natural killer (NK) cell populations, and therefore has potential for potentiating adoptive immune cell therapy for cancer. Previously, IL-15 has been shown to induce proliferation of CD8 memory T cells through activation of telomerase. Here, we investigated whether telomerase is also activated during the IL-15 mediated proliferation of NK and NKT-like (CD56+CD3+) cells. We also examined the extent that each of the three signaling pathways known to be stimulated by IL-2/IL-15 (JAK-STAT, PI3K-AKT Ras-RAF/MAPK) were activated and involved in the telomerase expression in the three cell types NK, NKT, or CD8 T cells. To assess cell proliferation and doubling, peripheral blood mononuclear cells (PBMCs) or isolated NK, NKT-like or CD8 T cells were incubated with varying concentrations of IL-15 or IL-2 for 7 days. CD8 T, NK, and NKT cell expansion was determined by fluorophore-conjugated antibody staining and flow cytometry. Cell doubling was investigated using carboxyfluorescein-succinimidyl-ester (CFSE). Telomerase expression was investigated by staining cells with anti-telomerase reverse transcriptase (anti-TERT). Telomerase activity in CD56+ and CD8 T cells was also measured via Telomerase Repeat Amplification Protocol (TRAP). Analysis of cellular expansion, proliferation and TERT expression concluded that IL-15 increased cellular growth of NK, NKT, and CD8 T cells more effectively than IL-2 using low or high doses. IL-15, increased TERT expression in NK and NKT cells by up to 2.5 fold, the same increase seen in CD8 T cells. IL-2 had effects on TERT expression only at high doses (100–1000 ng/ml). Proteome profiling identified that IL-15 activated selected signaling proteins in the three pathways (JAK-STAT, PI3K-AKT, Ras-MAPK) known to mediate IL-2/IL-15 signaling, more strongly than IL-2. Evaluation by signaling pathway inhibitors revealed that JAK/STAT and PI3K/AKT pathways are important in IL-15’s ability to upregulate TERT expression in NK and NKT cells, whereas all three pathways were involved in CD8 T cell TERT expression. In conclusion, this study shows that IL-15 potently stimulates TERT upregulation in NK and NKT cells in addition to CD8 T cells and is therefore a valuable tool for adoptive cell therapies.
Collapse
Affiliation(s)
- Fiona Watkinson
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Sandeep Krishan Nayar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Aradhana Rani
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Christina A Sakellariou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Oussama Elhage
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom.,Urology Centre, Guy's Hospital, London, United Kingdom
| | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom.,Urology Centre, Guy's Hospital, London, United Kingdom
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
12
|
Dratwa M, Wysoczańska B, Łacina P, Kubik T, Bogunia-Kubik K. TERT-Regulation and Roles in Cancer Formation. Front Immunol 2020; 11:589929. [PMID: 33329574 PMCID: PMC7717964 DOI: 10.3389/fimmu.2020.589929] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is a catalytic subunit of telomerase. Telomerase complex plays a key role in cancer formation by telomere dependent or independent mechanisms. Telomere maintenance mechanisms include complex TERT changes such as gene amplifications, TERT structural variants, TERT promoter germline and somatic mutations, TERT epigenetic changes, and alternative lengthening of telomere. All of them are cancer specific at tissue histotype and at single cell level. TERT expression is regulated in tumors via multiple genetic and epigenetic alterations which affect telomerase activity. Telomerase activity via TERT expression has an impact on telomere length and can be a useful marker in diagnosis and prognosis of various cancers and a new therapy approach. In this review we want to highlight the main roles of TERT in different mechanisms of cancer development and regulation.
Collapse
Affiliation(s)
- Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Barbara Wysoczańska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Tomasz Kubik
- Department of Computer Engineering, Faculty of Electronics, Wrocław University of Science and Technology, Wroclaw, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
13
|
Zhai L, Zhao Y, Liu Z, Wu J, Lin L. mRNA expression profile analysis reveals a C-MYC/miR-34a pathway involved in the apoptosis of diffuse large B-cell lymphoma cells induced by Yiqichutan treatment. Exp Ther Med 2020; 20:2157-2165. [PMID: 32765691 PMCID: PMC7401774 DOI: 10.3892/etm.2020.8940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of adult non-Hodgkin's lymphoma (NHL). While DLBCL is sensitive to chemotherapy, a certain percentage of patients with DLBCL experience relapse. Previous studies have indicated that Yiqichutan treatment, which was developed to treat NHL, can inhibit DLBCL cell growth, but the mechanism is not fully understood. The present study identified 991 differentially expressed mRNAs, with 498 upregulated and 493 downregulated (P<0.05), in SUDHL-6 cells exposed to Yiqichutan. The underlying pathways included the Jak/Stat and PI3K signaling pathways. In total, six representative mRNAs were selected for validation with reverse transcription-quantitative PCR (RT-qPCR), and a strong correlation was identified between the RT-qPCR results and microarray data. Since the transcription factor C-MYC is involved in both the Jak/Stat and PI3K signaling pathways, C-MYC and its associated microRNA (miR) were selected for further analysis. It was found that knockdown of C-MYC increased miR-34a expression levels, inhibited forkhead box P1 (Foxp1) expression levels and promoted DLBCL cell apoptosis. In addition, the miR-34a mimics further enhanced the role of C-MYC knockdown. It was demonstrated that, the expression levels of apoptotic factors Bax and poly (ADP-ribose) polymerase were significantly upregulated with C-MYC knockdown and miR-34a mimics in SUDHL-6 cells, while the Bcl2 expression level was significantly reduced. Moreover, Yiqichutan treatment increased miR-34a expression levels and induced apoptosis, as well as reducing Foxp1 expression level in SUDHL-6 cells. Therefore, the present results suggested that Yiqichutan treatment affected DLBCL cells via several signaling pathways. Furthermore, Yiqichutan may inhibit the proliferation of DLBCL cells by blocking the C-MYC/miR-34a signaling pathway.
Collapse
Affiliation(s)
- Linzhu Zhai
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Zeyu Liu
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Jie Wu
- Project Department, Forevergen Biosciences Co., Ltd., Guangzhou, Guangdong 510000, P.R. China
| | - Lizhu Lin
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
14
|
Zhu XY, Guo QY, Zhu M, Chen BG, Wang LY, Zhang DQ, Zhang L, Shao YP, Luo WD. HLX affects cell cycle and proliferation in AML cells via the JAK/STAT signaling pathway. Oncol Lett 2020; 20:1888-1896. [PMID: 32724432 PMCID: PMC7377103 DOI: 10.3892/ol.2020.11718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/13/2020] [Indexed: 01/11/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a class of malignant tumors derived from hematopoietic stem or progenitor cells. The H2.0-like homeobox gene (HLX) encodes transcription factors that function in promoting normal hematopoietic cell proliferation and tumor immunity. The present study analyzed the effect of downregulating the HLX on cell cycle distribution and cell proliferation in AML. Moreover, the current study detected changes in the expression of genes and proteins in the Janus kinase (JAK)/STAT signaling pathway to investigate the mechanism of the action of HLX in tumor immunity in AML. HLX expression in AML cell lines was silenced using small interfering siRNA, and MTS/PMS-assay colorimetric assays were used to assess the effect of knockdown of HLX on AML cell proliferation. Flow cytometry was used to analyze changes in cell cycle distribution, while reverse transcription-quantitative PCR and western blotting were used to detect changes in the expression levels of key components of the JAK/STAT signaling pathway, such as p21-activated kinase 1 (PAK1), neuropilin 1 (NRP1), B-cell translocation gene 1 (BTG1) and STAT5. It was found that HLX was differentially expressed in AML cell lines of various subtypes, and HLX expression was higher in the AML/M3 subtype NB4 cell line compared with the control group. Knockdown of HLX in NB4 cells significantly inhibited cell proliferation and arrested cells in the G0/G1 phase. Moreover, STAT5 protein expression, as well as NRP1 and PAK1 expression levels were downregulated, while BTG1 expression was upregulated when HLX was knocked out by siRNA. Collectively, the results suggested that downregulation of HLX may cause G0/G1 phase arrest and inhibit the proliferation of AML cells by activating the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Xia-Yin Zhu
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Qun-Yi Guo
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Min Zhu
- Department of Central Laboratory, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Bao-Guo Chen
- Department of Central Laboratory, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Ling-Yan Wang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Dan-Qiong Zhang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Li Zhang
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Yan-Ping Shao
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| | - Wen-Da Luo
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical College, Taizhou, Zhejiang 317000, P.R. China
| |
Collapse
|
15
|
Majumder MM, Leppä AM, Hellesøy M, Dowling P, Malyutina A, Kopperud R, Bazou D, Andersson E, Parsons A, Tang J, Kallioniemi O, Mustjoki S, O'Gorman P, Wennerberg K, Porkka K, Gjertsen BT, Heckman CA. Multi-parametric single cell evaluation defines distinct drug responses in healthy hematologic cells that are retained in corresponding malignant cell types. Haematologica 2020; 105:1527-1538. [PMID: 31439679 PMCID: PMC7271564 DOI: 10.3324/haematol.2019.217414] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/22/2019] [Indexed: 01/22/2023] Open
Abstract
Innate drug sensitivity in healthy cells aids identification of lineage specific anti-cancer therapies and reveals off-target effects. To characterize the diversity in drug responses in the major hematopoietic cell types, we simultaneously assessed their sensitivity to 71 small molecules utilizing a multi-parametric flow cytometry assay and mapped their proteomic and basal signaling profiles. Unsupervised hierarchical clustering identified distinct drug responses in healthy cell subsets based on their cellular lineage. Compared to other cell types, CD19+/B and CD56+/NK cells were more sensitive to dexamethasone, venetoclax and midostaurin, while monocytes were more sensitive to trametinib. Venetoclax exhibited dose-dependent cell selectivity that inversely correlated to STAT3 phosphorylation. Lineage specific effect of midostaurin was similarly detected in CD19+/B cells from healthy, acute myeloid leukemia and chronic lymphocytic leukemia samples. Comparison of drug responses in healthy and neoplastic cells showed that healthy cell responses are predictive of the corresponding malignant cell response. Taken together, understanding drug sensitivity in the healthy cell-of-origin provides opportunities to obtain a new level of therapy precision and avoid off-target toxicity.
Collapse
Affiliation(s)
- Muntasir M Majumder
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Aino-Maija Leppä
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Monica Hellesøy
- Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - Paul Dowling
- Department of Biology, National University of Ireland, Maynooth, Ireland
| | - Alina Malyutina
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Reidun Kopperud
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Despina Bazou
- Department of Hematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Emma Andersson
- Department of Clinical Chemistry and Hematology, University of Helsinki, Finland
| | - Alun Parsons
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jing Tang
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Solna, Sweden
| | - Satu Mustjoki
- Department of Clinical Chemistry and Hematology, University of Helsinki, Finland
- Hematology Research Unit Helsinki, University of Helsinki, Helsinki, Finland
| | - Peter O'Gorman
- Department of Hematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- BRIC-Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Kimmo Porkka
- Hematology Research Unit Helsinki, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Bjørn T Gjertsen
- Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Dahlström J, Xia C, Xing X, Yuan X, Björkholm M, Xu D. JAK2 inhibition in JAK2 V617F-bearing leukemia cells enriches CD34 + leukemic stem cells that are abolished by the telomerase inhibitor GRN163L. Biochem Biophys Res Commun 2020; 527:425-431. [PMID: 32334833 DOI: 10.1016/j.bbrc.2020.04.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
The activating-mutation of JAK2V617F drives the development of myeloproliferative neoplasms (MPNs). Several JAK2 inhibitors such as ruxolitinib and gandotinib (LY2784544) currently in clinical trials and, provide improvements in MPNs including myelofibrosis. However, JAK2 inhibitors are non-curative and murine experiments show that JAK2 inhibitors don't eradicate MPN stem cells and it is currently unclear how they escape. We thus determined the effect of the specific JAK2V617F inhibitor LY2784544 on leukemic stem (CD34+) cells (LSCs) using the JAK2V617F-bearing erythroleukemia cell line HEL. The LY2784544 treatment caused a transient proliferation inhibition and apoptosis of HEL cells, but a recovery occurred within a week. Thereafter, the continuous LY2784544 exposure induced the accumulation of CD34+ LSCs, and the CD34+ cells increased from 2% to >90% by week 9, which was accompanied by increased clonogenic potentials. LY2784544 was capable of stimulating CD34 expression even in CD34- HEL cells, which indicated cellular de-differentiation. A significantly enhanced expression of the stem cell factor KLF4 was observed in LY2784544-treated HEL cells. Inhibiting KLF4 expression attenuated LY2784544-mediated accumulation of CD34+ LSCs. Moreover, the telomerase inhibitor GRN163L abolished the LY2784544-effect. JAK2 inhibitors thus cause enrichment of LSCs and are unlikely to cure MPN as a monotherapy. Simultaneously targeting JAK2V617F and KLF4 or telomerase may be a novel strategy for MPN therapy, which should be of significance both biologically and clinically.
Collapse
Affiliation(s)
- Jenny Dahlström
- Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Chuanyou Xia
- Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Xiangling Xing
- Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Xiaotian Yuan
- School of Medicine, Shandong University, Jinan, PR China.
| | - Magnus Björkholm
- Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
17
|
Yu S, Xu T, Dai J, Ma M, Tang H, Chi Z, Si L, Cui C, Sheng X, Kong Y, Guo J. TERT copy gain predicts the outcome of high-dose interferon α-2b therapy in acral melanoma. Onco Targets Ther 2018; 11:4097-4104. [PMID: 30046245 PMCID: PMC6054280 DOI: 10.2147/ott.s158239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Asian populations are more likely to develop acral melanoma (AM) than Caucasians. The high-dose interferon (HD-IFN) α-2b regimen is the main adjuvant treatment for AM. TERT encodes the catalytic subunit of telomerase reverse transcriptase, which plays an important role in melanoma. Frequent TERT mutation and increased TERT gene expression have been described in AM. Our study aimed to investigate the status and the clinical significance of TERT copy number in a large cohort of patients with AM and to analyze the relationship between TERT copy number gain and the efficiency of HD-IFN. Patients and methods A total of 573 melanoma samples were retrospectively collected and analyzed for TERT copy number via Sanger sequencing. Clinical data of patients were also collected. Results TERT copy gain (copy number >2) was detected in 257 of the 573 patients with AM (44.9%). Of the 573 patients, 81 (14.1%) had a high copy gain (copy number >4). Patients with ulceration showed a significantly higher copy gain rate of TERT compared to the patients without ulceration (P=0.028). Patients with a tumor thicker than 4 mm also had a higher copy number rate of TERT than those with <4 mm (P=0.048). Our results showed that the overall survival (OS) was not significantly different between patients with and without TERT copy gain (P=0.890). However, among the 278 patients who received an HD-IFN regimen, Kaplan–Meier survival analysis demonstrated a significant correlation between TERT copy gain and relapse-free survival (RFS) (P=0.008). In addition, multivariate Cox regression assays validated TERT copy gain to be an independent prognostic factor of RFS for patients with AM undergoing HD-IFN therapy (hazard ratio =1.50; P=0.019). Conclusion The copy number status of TERT might be a predictor for HD-IFN efficacy, but it is not a prognostic factor of OS in patients with AM.
Collapse
Affiliation(s)
- Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing 100142, China, ;
| |
Collapse
|
18
|
Targeting PFKFB3 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitor. Oncogene 2018; 37:2837-2849. [DOI: 10.1038/s41388-018-0157-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 01/10/2018] [Accepted: 01/14/2018] [Indexed: 01/20/2023]
|
19
|
Abstract
Length of the telomere (TL), a structure at the tip of chromosome that protects and ensures stability, is determined by multi-protein complexes such as telosome/shelterin and telomerase. Earlier studies from our laboratory show that longer TL has potential to be positive predictive biomarker of clinical outcome to anti-epidermal growth factor receptor (EGFR) monoclonal antibody therapy in patients with KRAS WT metastatic colorectal cancer. Although there is extensive literature suggesting the role of shelterin and telomerase, not much literature exists that describes the role of EGFR and KRAS pathway in regulating TL. This detailed review focuses on an insight into various components, including proteins, enzymes and transcription factors, interlinking between EGFR pathways and telomerase that regulate TL.
Collapse
|
20
|
Beldi-Ferchiou A, Skouri N, Ben Ali C, Safra I, Abdelkefi A, Ladeb S, Mrad K, Ben Othman T, Ben Ahmed M. Abnormal repression of SHP-1, SHP-2 and SOCS-1 transcription sustains the activation of the JAK/STAT3 pathway and the progression of the disease in multiple myeloma. PLoS One 2017; 12:e0174835. [PMID: 28369102 PMCID: PMC5378363 DOI: 10.1371/journal.pone.0174835] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
Sustained activation of JAK/STAT3 signaling pathway is classically described in Multiple Myeloma (MM). One explanation could be the silencing of the JAK/STAT suppressor genes, through the hypermethylation of SHP-1 and SOCS-1, previously demonstrated in MM cell lines or in whole bone marrow aspirates. The link between such suppressor gene silencing and the degree of bone marrow invasion or the treatment response has not been evaluated in depth. Using real-time RT-PCR, we studied the expression profile of three JAK/STAT suppressor genes: SHP-1, SHP-2 and SOCS-1 in plasma cells freshly isolated from the bone marrows of MM patients and healthy controls. Our data demonstrated an abnormal repression of such genes in malignant plasma cells and revealed a significant correlation between such defects and the sustained activation of the JAK/STAT3 pathway during MM. The repressed expression of SHP-1 and SHP-2 correlated significantly with a high initial degree of bone marrow infiltration but was, unexpectedly, associated with a better response to the induction therapy. Collectively, our data provide new evidences that substantiate the contribution of JAK/STAT suppressor genes in the pathogenesis of MM. They also highlight the possibility that the decreased gene expression of SHP-1 and SHP-2 could be of interest as a new predictive factor of a favorable treatment response, and suggest new potential mechanisms of action of the therapeutic molecules. Whether such defect helps the progression of the disease from monoclonal gammopathy of unknown significance to MM remains, however, to be determined.
Collapse
Affiliation(s)
- Asma Beldi-Ferchiou
- Institut Pasteur de Tunis, Laboratory of Clinical Immunology, Tunis, Tunisia
| | - Nour Skouri
- Institut Pasteur de Tunis, Laboratory of Clinical Immunology, Tunis, Tunisia
| | - Cyrine Ben Ali
- Institut Pasteur de Tunis, Laboratory of Clinical Immunology, Tunis, Tunisia
| | - Ines Safra
- Institut Pasteur de Tunis, Laboratory of Molecular and Cellular Hematology, Tunis, Tunisia
- Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunis, Tunisie
| | | | - Saloua Ladeb
- Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunis, Tunisie
- Bone Marrow Transplantation Center, Tunis, Tunisia
| | - Karima Mrad
- Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunis, Tunisie
- Salah Azaiez Institute, Department of Pathology, Tunis, Tunisia
| | - Tarek Ben Othman
- Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunis, Tunisie
- Bone Marrow Transplantation Center, Tunis, Tunisia
| | - Mélika Ben Ahmed
- Institut Pasteur de Tunis, Laboratory of Clinical Immunology, Tunis, Tunisia
- Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunis, Tunisie
- * E-mail:
| |
Collapse
|
21
|
De Kouchkovsky I, Abdul-Hay M. 'Acute myeloid leukemia: a comprehensive review and 2016 update'. Blood Cancer J 2016; 6:e441. [PMID: 27367478 PMCID: PMC5030376 DOI: 10.1038/bcj.2016.50] [Citation(s) in RCA: 881] [Impact Index Per Article: 97.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/03/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with an incidence of over 20 000 cases per year in the United States alone. Large chromosomal translocations as well as mutations in the genes involved in hematopoietic proliferation and differentiation result in the accumulation of poorly differentiated myeloid cells. AML is a highly heterogeneous disease; although cases can be stratified into favorable, intermediate and adverse-risk groups based on their cytogenetic profile, prognosis within these categories varies widely. The identification of recurrent genetic mutations, such as FLT3-ITD, NMP1 and CEBPA, has helped refine individual prognosis and guide management. Despite advances in supportive care, the backbone of therapy remains a combination of cytarabine- and anthracycline-based regimens with allogeneic stem cell transplantation for eligible candidates. Elderly patients are often unable to tolerate such regimens, and carry a particularly poor prognosis. Here, we review the major recent advances in the treatment of AML.
Collapse
Affiliation(s)
- I De Kouchkovsky
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - M Abdul-Hay
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Department of Hematology/Oncology, New York University Perlmutter Cancer Center, New York, NY, USA
| |
Collapse
|