1
|
Xu P, Sun X, Pan L, Zhu J, Qian S. Disulfidptosis-related LncRNAs forecast the prognosis of acute myeloid leukemia. Sci Rep 2025; 15:13635. [PMID: 40254646 PMCID: PMC12009979 DOI: 10.1038/s41598-025-95607-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy with a poor prognosis for patients. Disulfidptosis response-related long non-coding RNAs (DRLs) have been demonstrated to be closely associated with cancer development. Therefore, this study aims to construct a prognostic DRL signature and investigate the immune microenvironment for AML. RNA-seq and clinical data for AML patients were obtained from The Cancer Genome Atlas (TCGA) database. A total of 344 disulfidptosis-associated lncRNAs were identified, and a prognostic model consisting of seven lncRNAs was constructed and validated. Two risk groups, high-risk and low-risk, were identified. The model demonstrated a robust capacity to predict prognosis, with a worse overall survival for patients in the high-risk group. Additionally, differential expression of the seven lncRNAs were relatively higher in AML samples than in control samples via quantitative polymerase chain reaction(qPCR). The Kyoto Encyclopedia of Genes and Genomes (KEGG) and immune infiltration analysis revealed a substantial infiltration of immune cells and enrichment of immune pathways in the high-risk group. The sensitivity of AML patients to drugs varied according to their risk grade. This study identified a DRL signature, which can effectively predict the prognosis of AML and better understand the mechanism of disulfidptosis in AML. This provides a basis for personalized immunotherapy in AML patients.
Collapse
Affiliation(s)
- Pei Xu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Xiaolin Sun
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Lingxiao Pan
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Jianfeng Zhu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Sixuan Qian
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
| |
Collapse
|
2
|
Wang B, Jiang H, Sun N, Wang Z, Wang C, Yang T, Wang Y, Wang L. Angelica sinensis polysaccharides ameliorate 5-FU-induced stress anemia via promoting extramedullary erythroblastic island central macrophage-mediated erythroid differentiation. Int Immunopharmacol 2024; 142:113061. [PMID: 39260313 DOI: 10.1016/j.intimp.2024.113061] [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: 03/03/2024] [Revised: 07/30/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Chronic anemia, especially chemotherapy-induced anemia, is a common and intractable symptom. Puzzlingly, the conventional anemic treatment may lead to various side effects, and the mechanism of stress anemia remains unclear. METHODS Here, peripheral blood, histopathological and transmission electron microscopical examination, colony forming test, flow cytometry, and qRT-PCR assay were used to investigate the effects of Angelia sinensis polysaccharide (ASP), one main active ingredient of Chinese herb medicine Angelica sinensis, on ameliorating 5-fluorouracil (5-FU)-induced stress anemia. RESULTS We found that intraperitoneal injection to a C57BL/6J mouse ASP 100 mg/kg per day for consecutive 10 days or 14 days, remarkably accelerated the recovery of RBC, hemoglobin, and hematocrit in blood. ASP alleviated 5-FU-caused impairment of bone marrow cell and BFU-E enumeration. Meanwhile, ASP antagonized 5-FU promoting extramedullary erythropoiesis in the spleen, inducing splenomegaly due to stress erythroblastic islands, and occurrence of megakaryocytes and hematopoietic precursors in splenic colonies. ASP increased splenic stress BFU-E enumeration, driving BFU-E differentiation towards Pro-E and end-stage erythroblasts. Furthermore, ASP increased the number of F4/80+VCAM-1+ splenic erythroblastic island central macrophages, upregulating genetic expression of EPOR, Emp, VCAM-1, Hmox-1, Trf, TfR1, Fpn1, Spi-C, DNase2a, Tim4, MertK, and Klf1 in splenocytes. CONCLUSIONS Our findings indicate that the possible mechanism of chemotherapy-induced anemia is related to stress erythroid maturation arrest. Whereas, ASP may promote stress erythroid differentiation via elevated EPO sensitivity in extramedullary hematopoietic organs and enhanced macrophage-mediated adhesion, iron homeostasis and transfer, and nuclear engulfment, which may represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Biyao Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Honghui Jiang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Nianci Sun
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ziling Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Cheng Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ting Yang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Lu Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
3
|
Rodriguez-Sevilla JJ, Colla S. Inflammation in myelodysplastic syndrome pathogenesis. Semin Hematol 2024; 61:385-396. [PMID: 39424469 DOI: 10.1053/j.seminhematol.2024.09.005] [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: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Inflammation is a key driver of the progression of preleukemic myeloid conditions, such as clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS), to myelodysplastic syndromes (MDS). Inflammation is a critical mediator in the complex interplay of the genetic, epigenetic, and microenvironmental factors contributing to clonal evolution. Under inflammatory conditions, somatic mutations in TET2, DNMT3A, and ASXL1, the most frequently mutated genes in CHIP and CCUS, induce a competitive advantage to hematopoietic stem and progenitor cells, which leads to their clonal expansion in the bone marrow. Chronic inflammation also drives metabolic reprogramming and immune system deregulation, further promoting the expansion of malignant clones. This review underscores the urgent need to fully elucidate the role of inflammation in MDS initiation and highlights the potential of the therapeutical targeting of inflammatory pathways as an early intervention in MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
4
|
Choi HS, Kim BS, Yoon S, Oh SO, Lee D. Leukemic Stem Cells and Hematological Malignancies. Int J Mol Sci 2024; 25:6639. [PMID: 38928344 PMCID: PMC11203822 DOI: 10.3390/ijms25126639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
The association between leukemic stem cells (LSCs) and leukemia development has been widely established in the context of genetic alterations, epigenetic pathways, and signaling pathway regulation. Hematopoietic stem cells are at the top of the bone marrow hierarchy and can self-renew and progressively generate blood and immune cells. The microenvironment, niche cells, and complex signaling pathways that regulate them acquire genetic mutations and epigenetic alterations due to aging, a chronic inflammatory environment, stress, and cancer, resulting in hematopoietic stem cell dysregulation and the production of abnormal blood and immune cells, leading to hematological malignancies and blood cancer. Cells that acquire these mutations grow at a faster rate than other cells and induce clone expansion. Excessive growth leads to the development of blood cancers. Standard therapy targets blast cells, which proliferate rapidly; however, LSCs that can induce disease recurrence remain after treatment, leading to recurrence and poor prognosis. To overcome these limitations, researchers have focused on the characteristics and signaling systems of LSCs and therapies that target them to block LSCs. This review aims to provide a comprehensive understanding of the types of hematopoietic malignancies, the characteristics of leukemic stem cells that cause them, the mechanisms by which these cells acquire chemotherapy resistance, and the therapies targeting these mechanisms.
Collapse
Affiliation(s)
- Hee-Seon Choi
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
5
|
Fan C, Yang X, Yan L, Shi Z. Oxidative stress is two-sided in the treatment of acute myeloid leukemia. Cancer Med 2024; 13:e6806. [PMID: 38715546 PMCID: PMC11077289 DOI: 10.1002/cam4.6806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Oxidative stress caused by elevated ROS, as a novel therapeutic mechanism, has been implicated in various tumors including AML. AML cells are chronically under oxidative stress, yet overreliance on ROS production makes tumor cells increasingly vulnerable to further damage. Reducing the cytotoxic effect of ROS on normal cells while killing leukemia stem cell (LSC) with high levels of reactive oxygen species is a new challenge for oxidative stress therapy in leukemia. METHODS By searching literature databases, we summarized recent relevant studies. The relationship of ROS on AML genes, signaling pathways, and transcription factors, and the correlation of ROS with AML bone marrow microenvironment and autophagy were summarized. In addition, we summarize the current status of research on ROS and AML therapeutics. Finally, we discuss the research progress on redox resistance in AML. RESULTS This review discusses the evidence showing the link between redox reactions and the progression of AML and compiles the latest research findings that will facilitate future biological studies of redox effects associated with AML treatment. CONCLUSION We believe that exploiting this unique oxidative stress property of AML cells may provide a new way to prevent relapse and drug resistance.
Collapse
Affiliation(s)
- Chenyang Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Xiangdong Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Lixiang Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Zhexin Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| |
Collapse
|
6
|
Khorashad JS, Rizzo S, Tonks A. Reactive oxygen species and its role in pathogenesis and resistance to therapy in acute myeloid leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:5. [PMID: 38434766 PMCID: PMC10905166 DOI: 10.20517/cdr.2023.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Relapse following a short clinical response to therapy is the major challenge for the management of acute myeloid leukemia (AML) patients. Leukemic stem cells (LSC), as the source of relapse, have been investigated for their metabolic preferences and their alterations at the time of relapse. As LSC rely on oxidative phosphorylation (OXPHOS) for energy requirement, reactive oxygen species (ROS), as by-products of OXPHOS, have been investigated for their role in the effectiveness of the standard AML therapy. Increased levels of non-mitochondrial ROS, generated by nicotinamide adenine dinucleotide phosphate oxidase, in a subgroup of AML patients add to the complexity of studying ROS. Although there are various studies presenting the contribution of ROS to AML pathogenesis, resistance, and its inhibition or activation as a target, a model that can clearly explain its role in AML has not been conceptualized. This is due to the heterogeneity of AML, the dynamics of ROS production, which is influenced by factors such as the type of treatment, cell differentiation state, mitochondrial activity, and also the heterogeneous generation of non-mitochondrial ROS and limited available data on their interaction with the microenvironment. This review summarizes these challenges and the recent progress in this field.
Collapse
Affiliation(s)
- Jamshid Sorouri Khorashad
- Department of Immunology and inflammation, Imperial College London, London, W12 0NN, UK
- Department of Molecular Pathology, Institute of Cancer Research, Sutton, SM2 5PT, UK
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sian Rizzo
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alex Tonks
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
7
|
Nguyen TM, Jang WB, Lee Y, Kim YH, Lim HJ, Lee EJ, Nguyen TMT, Choi EJ, Kwon SM, Oh JW. Non-intrusive quality appraisal of differentiation-induced cardiovascular stem cells using E-Nose sensor technology. Biosens Bioelectron 2024; 246:115838. [PMID: 38042052 DOI: 10.1016/j.bios.2023.115838] [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: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/11/2023] [Indexed: 12/04/2023]
Abstract
Stem cell technology holds immense potential for revolutionizing medicine, particularly in regenerative treatment for heart disease. The unique capacity of stem cells to differentiate into diverse cell types offers promise in repairing damaged tissues and implanting organs. Ensuring the quality of differentiated cells, essential for specific functions, demands in-depth analysis. However, this process consumes time and incurs substantial costs while invasive methods may alter stem cell features during differentiation and deplete cell numbers. To address these challenges, we propose a non-invasive strategy, using cellular respiration, to assess the quality of differentiation-induced stem cells, notably cardiovascular stem cells. This evaluation employs an electronic nose (E-Nose) and neural pattern separation (NPS). Our goal is to assess differentiation-induced cardiac stem cells (DICs) quality through E-Nose data analysis and compare it with standard commercial human cells (SCHCs). Sensitivity and specificity were evaluated by interacting SCHCs and DICs with the E-Nose, achieving over 90% classification accuracy. Employing selective combinations optimized by NPS, E-Nose successfully classified all six cell types. Consequently, the relative similarity among DICs like cardiomyocytes, endothelial cells with SCHCs was established relied on comparing response data from the E-Nose sensor without resorting to complex evaluations.
Collapse
Affiliation(s)
- Thanh Mien Nguyen
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Yujin Lee
- Department of Nano Fusion Technology, Pusan National University, Busan, 46214, Republic of Korea
| | - You Hwan Kim
- Department of Nano Fusion Technology, Pusan National University, Busan, 46214, Republic of Korea
| | - Hye Ji Lim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Thu M T Nguyen
- Department of Nano Fusion Technology, Pusan National University, Busan, 46214, Republic of Korea
| | - Eun-Jung Choi
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea.
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Jin-Woo Oh
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, Republic of Korea; Department of Nano Fusion Technology, Pusan National University, Busan, 46214, Republic of Korea.
| |
Collapse
|
8
|
Farrag EK, Aziz WM, Shaker SE, Shawky H, Fayed DB. Toxicological profiling of a de novo synthesized benzimidazole derivative with potent and selective proapoptotic potentials against breast cancer. Food Chem Toxicol 2023; 180:114049. [PMID: 37734466 DOI: 10.1016/j.fct.2023.114049] [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: 07/26/2023] [Revised: 08/19/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
This study aimed to investigate the toxicological profile of 1-(6-(1H-benzo[d]imidazole-2-yl)-2-methylpyridin-3-yl) ethanone (BMPE), both in vitro and in vivo. The proapoptotic/necrotic and cell cycle arrest potentials of BMPE were assessed in MCF-7 cell line. The in vivo toxicology was assessed in female Balb/c mice by repeated dosing of 5, 25, and 50 mg/kg for 21 consecutive days, then different biochemical, inflammatory, and oxidative markers were assessed in sera/tissue homogenates of treated animals. The new derivative showed a potent selective cytotoxicity against malignant cell lines with IC50 value 0.2 μM/mL, while the cytotoxic effect on normal Wi-38 cells was observed at IC50 value 0.4 μM/mL; i.e. twofold the effective anticancer dose. BMPE exhibited an early DNA fragmentation-derived cell apoptosis observed at the G0/G1 checkpoint. In vivo, BMPE was biochemically/immunologically tolerable at a pharmacological dose range of 5-25 mg/kg, with no significant rates of mortality/morbidity and minimal-to-moderate histopathological alterations recorded. The new derivative represents an attractive therapeutic candidate for breast cancer, considering its noticeable modulatory effect on the oxidative-inflammatory axis that would relate to its potent antitumor effect.
Collapse
Affiliation(s)
- Ebtehal K Farrag
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Wessam M Aziz
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Sylvia E Shaker
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Heba Shawky
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622, Cairo, Egypt.
| | - Dalia B Fayed
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki, 12622, Cairo, Egypt
| |
Collapse
|
9
|
Adema V, Ganan-Gomez I, Ma F, Rodriguez-Sevilla JJ, Chien K, Yang H, Thongon N, Kanagal-Shamanna R, Loghavi S, Montalban-Bravo G, Hammond D, Gu Y, Tan R, Tan L, Lorenzi P, Al-Atrash G, Clise-Dwyer K, Bejar R, Pellegrini M, Garcia-Manero G, Colla S. IL-1β-mediated inflammatory signaling drives ineffective erythropoiesis in early-stage myelodysplastic syndromes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.560018. [PMID: 37808770 PMCID: PMC10557725 DOI: 10.1101/2023.09.28.560018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Myelodysplastic syndromes (MDS) are a group of incurable hematopoietic stem cell (HSC) neoplasms characterized by peripheral blood cytopenias and a high risk of progression to acute myeloid leukemia. MDS represent the final stage in a continuum of HSCs' genetic and functional alterations and are preceded by a premalignant phase, clonal cytopenia of undetermined significance (CCUS). Dissecting the mechanisms of CCUS maintenance may uncover therapeutic targets to delay or prevent malignant transformation. Here, we demonstrate that DNMT3A and TET2 mutations, the most frequent mutations in CCUS, induce aberrant HSCs' differentiation towards the myeloid lineage at the expense of erythropoiesis by upregulating IL-1β-mediated inflammatory signaling and that canakinumab rescues red blood cell transfusion dependence in early-stage MDS patients with driver mutations in DNMT3A and TET2 . This study illuminates the biological landscape of CCUS and offers an unprecedented opportunity for MDS intervention during its initial phase, when expected survival is prolonged.
Collapse
|
10
|
Pourcelot E, El Samra G, Mossuz P, Moulis JM. Molecular Insight into Iron Homeostasis of Acute Myeloid Leukemia Blasts. Int J Mol Sci 2023; 24:14307. [PMID: 37762610 PMCID: PMC10531764 DOI: 10.3390/ijms241814307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Acute myeloid leukemia (AML) remains a disease of gloomy prognosis despite intense efforts to understand its molecular foundations and to find efficient treatments. In search of new characteristic features of AML blasts, we first examined experimental conditions supporting the amplification of hematological CD34+ progenitors ex vivo. Both AML blasts and healthy progenitors heavily depended on iron availability. However, even if known features, such as easier engagement in the cell cycle and amplification factor by healthy progenitors, were observed, multiplying progenitors in a fully defined medium is not readily obtained without modifying their cellular characteristics. As such, we measured selected molecular data including mRNA, proteins, and activities right after isolation. Leukemic blasts showed clear signs of metabolic and signaling shifts as already known, and we provide unprecedented data emphasizing disturbed cellular iron homeostasis in these blasts. The combined quantitative data relative to the latter pathway allowed us to stratify the studied patients in two sets with different iron status. This categorization is likely to impact the efficiency of several therapeutic strategies targeting cellular iron handling that may be applied to eradicate AML blasts.
Collapse
Affiliation(s)
- Emmanuel Pourcelot
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX a9, France;
| | - Ghina El Samra
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
| | - Pascal Mossuz
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX a9, France;
- Team “Epigenetic and Cellular Signaling”, Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
| | - Jean-Marc Moulis
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), University Grenoble Alpes, INSERM U1055, 38000 Grenoble, France; (E.P.); (G.E.S.)
- University Grenoble Alpes, CEA, IRIG, 38000 Grenoble, France
| |
Collapse
|
11
|
Cancemi G, Cicero N, Allegra A, Gangemi S. Effect of Diet and Oxidative Stress in the Pathogenesis of Lymphoproliferative Disorders. Antioxidants (Basel) 2023; 12:1674. [PMID: 37759977 PMCID: PMC10525385 DOI: 10.3390/antiox12091674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Lymphomas are a heterogeneous group of pathologies that result from clonal proliferation of lymphocytes. They are classified into Hodgkin lymphoma and non-Hodgkin lymphoma; the latter develops as a result of B, T, or NK cells undergoing malignant transformation. It is believed that diet can modulate cellular redox state and that oxidative stress is implicated in lymphomagenesis by acting on several biological mechanisms; in fact, oxidative stress can generate a state of chronic inflammation through the activation of various transcription factors, thereby increasing the production of proinflammatory cytokines and causing overstimulation of B lymphocytes in the production of antibodies and possible alterations in cellular DNA. The purpose of our work is to investigate the results of in vitro and in vivo studies on the possible interaction between lymphomas, oxidative stress, and diet. A variety of dietary regimens and substances introduced with the diet that may have antioxidant and antiproliferative effects were assessed. The possibility of using nutraceuticals as novel anticancer agents is discussed; although the use of natural substances in lymphoma therapy is an interesting field of study, further studies are needed to define the efficacy of different nutraceuticals before introducing them into clinical practice.
Collapse
Affiliation(s)
- Gabriella Cancemi
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (G.C.); (A.A.)
| | - Nicola Cicero
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (G.C.); (A.A.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
12
|
Saulle E, Spinello I, Quaranta MT, Labbaye C. Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting. Cells 2023; 12:1553. [PMID: 37296673 PMCID: PMC10252746 DOI: 10.3390/cells12111553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Autophagy is a highly conserved cellular degradation process that regulates cellular metabolism and homeostasis under normal and pathophysiological conditions. Autophagy and metabolism are linked in the hematopoietic system, playing a fundamental role in the self-renewal, survival, and differentiation of hematopoietic stem and progenitor cells, and in cell death, particularly affecting the cellular fate of the hematopoietic stem cell pool. In leukemia, autophagy sustains leukemic cell growth, contributes to survival of leukemic stem cells and chemotherapy resistance. The high frequency of disease relapse caused by relapse-initiating leukemic cells resistant to therapy occurs in acute myeloid leukemia (AML), and depends on the AML subtypes and treatments used. Targeting autophagy may represent a promising strategy to overcome therapeutic resistance in AML, for which prognosis remains poor. In this review, we illustrate the role of autophagy and the impact of its deregulation on the metabolism of normal and leukemic hematopoietic cells. We report updates on the contribution of autophagy to AML development and relapse, and the latest evidence indicating autophagy-related genes as potential prognostic predictors and drivers of AML. We review the recent advances in autophagy manipulation, combined with various anti-leukemia therapies, for an effective autophagy-targeted therapy for AML.
Collapse
Affiliation(s)
- Ernestina Saulle
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| | | | | | - Catherine Labbaye
- Correspondence: (E.S.); (C.L.); Tel.: +39-0649902422 (E.S.); +39-0649902418 (C.L.)
| |
Collapse
|
13
|
Combined scRNAseq and Bulk RNAseq Analysis to Reveal the Dual Roles of Oxidative Stress-Related Genes in Acute Myeloid Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:5343746. [PMID: 36811020 PMCID: PMC9938912 DOI: 10.1155/2023/5343746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 02/11/2023]
Abstract
Background Oxidative stress (OS) can either lead to leukemogenesis or induce tumor cell death by inflammation and immune response accompanying the process of OS through chemotherapy. However, previous studies mainly focus on the level of OS state and the salient factors leading to tumorigenesis and progression of acute myeloid leukemia (AML), and nothing has been done to distinguish the OS-related genes with different functions. Method First, we downloaded single-cell RNA sequencing (scRNAseq) and bulk RNA sequencing (RNAseq) data from public databases and evaluated the oxidative stress functions between leukemia cells and normal cells by the ssGSEA algorithm. Then, we used machine learning methods to screen out OS gene set A related to the occurrence and prognosis of AML and OS gene set B related to treatment in leukemia stem cells (LSCs) like population (HSC-like). Furthermore, we screened out the hub genes in the above two gene sets and used them to identify molecular subclasses and construct a model for predicting therapy response. Results Leukemia cells have different OS functions compared to normal cells and significant OS functional changes before and after chemotherapy. Two different clusters in gene set A were identified, which showed different biological properties and clinical relevance. The sensitive model for predicting therapy response based on gene set B demonstrated predictive accuracy by ROC and internal validation. Conclusion We combined scRNAseq and bulk RNAseq data to construct two different transcriptomic profiles to reveal the different roles of OS-related genes involved in AML oncogenesis and chemotherapy resistance, which might provide important insights into the mechanism of OS-related genes in the pathogenesis and drug resistance of AML.
Collapse
|
14
|
Zhou H, Jiang Y, Huang Y, Zhong M, Qin D, Xie C, Pan G, Tan J, Deng M, Zhao H, Zhou Y, Tang Y, Lai Q, Fang Z, Luo Y, Jiang Y, Xu B, Zha J. Therapeutic inhibition of PPARα-HIF1α-PGK1 signaling targets leukemia stem and progenitor cells in acute myeloid leukemia. Cancer Lett 2023; 554:215997. [PMID: 36396101 DOI: 10.1016/j.canlet.2022.215997] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Treatment of acute myeloid leukemia (AML) with chemotherapeutic agents fails to eliminate leukemia stem cells (LSC),and thus patients remain at high risk for relapse. Therefore, the identification of agents that target LSC is an important consideration for the development of new therapies. Enhanced glycolysis in LSC contributes to the aggressiveness of AML, which is difficult to be targeted. In this study, we showed that targeting peroxisome-proliferator-activated receptor α (PPARα), a ligand-activated transcription factor by chiglitazar provided a promising therapeutic approach. We first identified that chiglitazar reduced cell viability and proliferation of the leukemia stem-like cells population in AML. Treatment with chiglitazar blocked the ubiquitination of PPARα and increased its expression, resulting in the inhibition of glucose metabolism and apoptosis of AML cells. Consistent with its anti-leukemia stem-like cells activity in vitro, chiglitazar treatment in vivo resulted in the significant killing of leukemia stem-like cells as demonstrated in AML patient-derived xenograft (PDX) models. Mechanistically, PPARα overexpression inhibited the expression and promoter activity of PGK1 through blocking HIF1-α interaction on the PGK1 promoter. Thus, we concluded that targeting PPARα may serve as a novel approach for enhancing stem and progenitor cells elimination in AML.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yuetin Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Mengya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Dongmei Qin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Chendi Xie
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guangchao Pan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Jinshui Tan
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Manman Deng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Haijun Zhao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yong Zhou
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yuanfang Tang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qian Lai
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yiming Luo
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China
| | - Yirong Jiang
- Department of Hematology, Affiliated Dongguan People's Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523059, China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China.
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China; Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, China.
| |
Collapse
|
15
|
A novel prognostic model of methylation-associated genes in acute myeloid leukemia. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1719-1728. [PMID: 36715873 DOI: 10.1007/s12094-022-03069-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/29/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND There is growing evidence that methylation-associated genes (MAGs) play an important role in the prognosis of acute myeloid leukemia (AML) patients. Thus, the aim of this research was to investigate the impact of MAGs in predicting the outcomes of AML patients. METHODS The expression profile and clinical information of patients were downloaded from public databases. A novel prognostic model based on 7 MAGs was established in the TCGA training cohort and validated in the GSE71014 dataset. To validate the clinical implications, the correlation between MAGs signature and drug sensitivity was further investigated. RESULTS 76 genes were screened out by the univariate Cox regression and significantly enriched in multiple methylation-related pathways. After filtering variables using LASSO regression analysis, 7 MAGs were introduced to construct the predictive model. The survival analysis showed overall survival of patients with the high-risk score was considerably poorer than that with the low-risk score in both the training and validating cohorts (p < 0.01). Furthermore, the risk score system as a prognostic factor also worked in the intermediate-risk patients based on ELN-2017 classification. Importantly, the risk score was demonstrated to be an independent prognostic factor for AML in the univariate and multivariate Cox regression analysis. Interestingly, GSEA analysis revealed that multiple metabolism-related pathways were significantly enriched in the high-risk group. Drug sensitivity analysis showed there was a significant difference in sensitivity of some drugs between the two groups. CONCLUSION We developed a robust and accurate prognostic model with 7 MAGs. Our findings might provide a reference for the clinical prognosis and management of AML.
Collapse
|
16
|
Identifying the Role of Oxidative Stress-Related Genes as Prognostic Biomarkers and Predicting the Response of Immunotherapy and Chemotherapy in Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6575534. [PMID: 36561981 PMCID: PMC9764017 DOI: 10.1155/2022/6575534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Background Ovarian cancer (OC) is one of the most frequently seen and fatal gynecological malignancies, and oxidative stress (OS) plays a critical role in the development and chemoresistance of OC. Materials and Methods OS-related genes (OSRGs) were obtained from the Molecular Signatures Database. Besides, gene expression profiles and clinical information from The Cancer Genome Atlas (TCGA) were selected to identify the prognostic OSRGs. Moreover, univariate Cox regression, LASSO, and multivariate Cox regression analyses were conducted sequentially to establish a prognostic signature, which was later validated in three independent Gene Expression Omnibus (GEO) datasets. Next, gene set enrichment analysis (GSEA) and tumor mutation burden (TMB) analysis were performed. Afterwards, immune checkpoint genes (ICGs) and the tumor immune dysfunction and exclusion (TIDE) algorithm, together with IMvigor210 and GSE78220 cohorts, were applied to comprehensively explore the role of OSRG signature in immunotherapy. Further, the CellMiner and Genomics of Drug Sensitivity in Cancer (GDSC) databases were also applied in investigating the significance of OSRG signature in chemotherapy. Results Altogether, 34 prognostic OSRGs were identified, among which 14 were chosen to establish the most valuable prognostic signature. The Kaplan-Meier (KM) analysis suggested that patients with lower OS-related risk score had better prognosis. The area under the curve (AUC) values were 0.71, 0.76, and 0.85 in 3, 5, and 7 years separately, and the stability of this prognostic signature was confirmed in three GEO datasets. As revealed by GSEA and TMB analysis results, OC patients in low-risk group might have better immunotherapeutic response, which was consistent with ICG expression and TIDE analyses. Moreover, both IMvigor210 and GSE78220 cohorts demonstrated that patients with lower OS-related risk score were more likely to benefit from anti-PD-1/L1 immunotherapy. In addition, the association between prognostic signature and drug sensitivity was explored. Conclusion According to our results in this work, OSRG signature can act as a powerful prognostic predictor for OC, which contributes to generating more individualized therapeutic strategies for OC patients.
Collapse
|
17
|
Arif T. Lysosomes and Their Role in Regulating the Metabolism of Hematopoietic Stem Cells. BIOLOGY 2022; 11:1410. [PMID: 36290314 PMCID: PMC9598322 DOI: 10.3390/biology11101410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSCs) have the capacity to renew blood cells at all stages of life and are largely quiescent at a steady state. It is essential to understand the processes that govern quiescence in HSCs to enhance bone marrow transplantation. It is hypothesized that in their quiescent state, HSCs primarily use glycolysis for energy production rather than mitochondrial oxidative phosphorylation (OXPHOS). In addition, the HSC switch from quiescence to activation occurs along a continuous developmental path that is driven by metabolism. Specifying the metabolic regulation pathway of HSC quiescence will provide insights into HSC homeostasis for therapeutic application. Therefore, understanding the metabolic demands of HSCs at a steady state is key to developing innovative hematological therapeutics. Lysosomes are the major degradative organelle in eukaryotic cells. Catabolic, anabolic, and lysosomal function abnormalities are connected to an expanding list of diseases. In recent years, lysosomes have emerged as control centers of cellular metabolism, particularly in HSC quiescence, and essential regulators of cell signaling have been found on the lysosomal membrane. In addition to autophagic processes, lysosomal activities have been shown to be crucial in sustaining quiescence by restricting HSCs access to a nutritional reserve essential for their activation into the cell cycle. Lysosomal activity may preserve HSC quiescence by altering glycolysis-mitochondrial biogenesis. The understanding of HSC metabolism has significantly expanded over the decade, revealing previously unknown requirements of HSCs in both their dividing (active) and quiescent states. Therefore, understanding the role of lysosomes in HSCs will allow for the development of innovative treatment methods based on HSCs to fight clonal hematopoiesis and HSC aging.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
18
|
Vilaplana-Lopera N, Cuminetti V, Almaghrabi R, Papatzikas G, Rout AK, Jeeves M, González E, Alyahyawi Y, Cunningham A, Erdem A, Schnütgen F, Raghavan M, Potluri S, Cazier JB, Schuringa JJ, Reed MAC, Arranz L, Günther UL, Garcia P. Crosstalk between AML and stromal cells triggers acetate secretion through the metabolic rewiring of stromal cells. eLife 2022; 11:e75908. [PMID: 36052997 PMCID: PMC9477493 DOI: 10.7554/elife.75908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
Acute myeloid leukaemia (AML) cells interact and modulate components of their surrounding microenvironment into their own benefit. Stromal cells have been shown to support AML survival and progression through various mechanisms. Nonetheless, whether AML cells could establish beneficial metabolic interactions with stromal cells is underexplored. By using a combination of human AML cell lines and AML patient samples together with mouse stromal cells and a MLL-AF9 mouse model, here we identify a novel metabolic crosstalk between AML and stromal cells where AML cells prompt stromal cells to secrete acetate for their own consumption to feed the tricarboxylic acid cycle (TCA) and lipid biosynthesis. By performing transcriptome analysis and tracer-based metabolic NMR analysis, we observe that stromal cells present a higher rate of glycolysis when co-cultured with AML cells. We also find that acetate in stromal cells is derived from pyruvate via chemical conversion under the influence of reactive oxygen species (ROS) following ROS transfer from AML to stromal cells via gap junctions. Overall, we present a unique metabolic communication between AML and stromal cells and propose two different molecular targets, ACSS2 and gap junctions, that could potentially be exploited for adjuvant therapy.
Collapse
Affiliation(s)
- Nuria Vilaplana-Lopera
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Vincent Cuminetti
- Stem Cells, Ageing and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT – The Arctic University of NorwayTromsoNorway
| | - Ruba Almaghrabi
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Department of Laboratory Medicine (hematology), Faculty of Applied Medical Sciences. Albaha University, Kingdom of Saudi ArabiaAl BahahSaudi Arabia
| | - Grigorios Papatzikas
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Computational Biology, University of BirminghamBirminghamUnited Kingdom
| | - Ashok Kumar Rout
- Institute of Chemistry and Metabolomics, University of LübeckLübeckGermany
| | - Mark Jeeves
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Elena González
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Yara Alyahyawi
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan UniversityJazanSaudi Arabia
| | - Alan Cunningham
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Ayşegül Erdem
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Frank Schnütgen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University FrankfurtFrankfurtGermany
- Frankfurt Cancer Institute, Goethe University FrankfurtFrankfurtGermany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, and German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Manoj Raghavan
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical CentreBirminghamUnited Kingdom
| | - Sandeep Potluri
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical CentreBirminghamUnited Kingdom
| | - Jean-Baptiste Cazier
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Computational Biology, University of BirminghamBirminghamUnited Kingdom
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Michelle AC Reed
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Lorena Arranz
- Stem Cells, Ageing and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT – The Arctic University of NorwayTromsoNorway
| | - Ulrich L Günther
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Institute of Chemistry and Metabolomics, University of LübeckLübeckGermany
| | - Paloma Garcia
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| |
Collapse
|
19
|
Targeting Redox Regulation as a Therapeutic Opportunity against Acute Leukemia: Pro-Oxidant Strategy or Antioxidant Approach? Antioxidants (Basel) 2022; 11:antiox11091696. [PMID: 36139768 PMCID: PMC9495346 DOI: 10.3390/antiox11091696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/07/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Redox adaptation is essential for human health, as the physiological quantities of non-radical reactive oxygen species operate as the main second messengers to regulate normal redox reactions by controlling several sensors. An abnormal increase reactive oxygen species, called oxidative stress, induces biological injury. For this reason, variations in oxidative stress continue to receive consideration as a possible approach to treat leukemic diseases. However, the intricacy of redox reactions and their effects might be a relevant obstacle; consequently, and alongside approaches aimed at increasing oxidative stress in neoplastic cells, antioxidant strategies have also been suggested for the same purpose. The present review focuses on the molecular processes of anomalous oxidative stress in acute myeloid and acute lymphoblastic leukemias as well as on the oxidative stress-determined pathways implicated in leukemogenic development. Furthermore, we review the effect of chemotherapies on oxidative stress and the possibility that their pharmacological effects might be increased by modifying the intracellular redox equilibrium through a pro-oxidant approach or an antioxidant strategy. Finally, we evaluated the prospect of varying oxidative stress as an efficacious modality to destroy chemoresistant cells using new methodologies. Altering redox conditions may be advantageous for inhibiting genomic variability and the eradication of leukemic clones will promote the treatment of leukemic disease.
Collapse
|
20
|
Machine Learning Assistants Construct Oxidative Stress-Related Gene Signature and Discover Potential Therapy Targets for Acute Myeloid Leukemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1507690. [PMID: 36046688 PMCID: PMC9423988 DOI: 10.1155/2022/1507690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 12/25/2022]
Abstract
Background Oxidative stress (OS) is associated with the development of acute myeloid leukemia (AML). However, there is lack of relevant research to confirm that OS-related genes can guide patients in risk stratification and predict their survival probability. Method First, we Data from three public databases, respectively. Then, we use batch univariate Cox regression and machine learning to select important characteristic genes; next, we build the model and use receiver operating characteristic curve (ROC) to evaluate the accuracy. Moreover, GSEAs were performed to discover the molecular mechanism and conduct nomogram visualization. In addition, the relative importance value was used to identify the hub gene, and GSE9476 was to validate hub gene difference expression. Finally, we use symptom mapping to predict the candidate herbs, targeting the hub gene, and put these candidate herbs into Traditional Chinese Medicine Systems Pharmacology (TCMSP) to identify the main small molecular ingredients and then docking hub proteins with this small molecular. Results A total of 313 candidate oxidative stress-related genes could affect patients' outcomes and machine learning to select six potential genes to construct a gene signature model to predict the overall survival (OS) of AML patients. Patients in a high group will obtain a short survival time when compared with the low-risk group (HR = 3.97, 95% CI: 2.48-6.36; p < 0.001). ROC results demonstrate the model has better prediction efficiency with AUC 0.873. GSEA suggests that this gene is enriched in several important signaling pathways. Nomogram is constructed and is robust. PLA2G4A is a hub gene of signature and associated with prognosis, and Nobiletin could target PLA2G4A for therapy AML. Conclusion We use two different machine learning methods to build six oxidative stress-related gene signatures that could assist clinical decisions and identify PLA2G4A as a potential biomarker for AML. Nobiletin, targeting PLA2G4, may provide a third pathway for therapy AML.
Collapse
|
21
|
Challenges in Cell Fate Acquisition to Scid-Repopulating Activity from Hemogenic Endothelium of hiPSCs Derived from AML Patients Using Forced Transcription Factor Expression. Cells 2022; 11:cells11121915. [PMID: 35741044 PMCID: PMC9221973 DOI: 10.3390/cells11121915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
The generation of human hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) represents a major goal in regenerative medicine and is believed would follow principles of early development. HSCs arise from a type of endothelial cell called a “hemogenic endothelium” (HE), and human HSCs are experimentally detected by transplantation into SCID or other immune-deficient mouse recipients, termed SCID-Repopulating Cells (SRC). Recently, SRCs were detected by forced expression of seven transcription factors (TF) (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1, and SPI1) in hPSC-derived HE, suggesting these factors are deficient in hPSC differentiation to HEs required to generate HSCs. Here we derived PECAM-1-, Flk-1-, and VE-cadherin-positive endothelial cells that also lack CD45 expression (PFVCD45−) which are solely responsible for hematopoietic output from iPSC lines reprogrammed from AML patients. Using HEs derived from AML patient iPSCs devoid of somatic leukemic aberrations, we sought to generate putative SRCs by the forced expression of 7TFs to model autologous HSC transplantation. The expression of 7TFs in hPSC-derived HE cells from an enhanced hematopoietic progenitor capacity was present in vitro, but failed to acquire SRC activity in vivo. Our findings emphasize the benefits of forced TF expression, along with the continued challenges in developing HSCs for autologous-based therapies from hPSC sources.
Collapse
|
22
|
Ye J, Calvo IA, Cenzano I, Vilas A, Martinez-de-Morentin X, Lasaga M, Alignani D, Paiva B, Viñado AC, San Martin-Uriz P, Romero JP, Quilez Agreda D, Miñana Barrios M, Sancho-González I, Todisco G, Malcovati L, Planell N, Saez B, Tegner JN, Prosper F, Gomez-Cabrero D. Deconvolution of the hematopoietic stem cell microenvironment reveals a high degree of specialization and conservation. iScience 2022; 25:104225. [PMID: 35494238 PMCID: PMC9046238 DOI: 10.1016/j.isci.2022.104225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/14/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022] Open
Abstract
Understanding the regulation of normal and malignant human hematopoiesis requires comprehensive cell atlas of the hematopoietic stem cell (HSC) regulatory microenvironment. Here, we develop a tailored bioinformatic pipeline to integrate public and proprietary single-cell RNA sequencing (scRNA-seq) datasets. As a result, we robustly identify for the first time 14 intermediate cell states and 11 stages of differentiation in the endothelial and mesenchymal BM compartments, respectively. Our data provide the most comprehensive description to date of the murine HSC-regulatory microenvironment and suggest a higher level of specialization of the cellular circuits than previously anticipated. Furthermore, this deep characterization allows inferring conserved features in human, suggesting that the layers of microenvironmental regulation of hematopoiesis may also be shared between species. Our resource and methodology is a stepping-stone toward a comprehensive cell atlas of the BM microenvironment.
Collapse
Affiliation(s)
- Jin Ye
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| | - Isabel A. Calvo
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Itziar Cenzano
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | - Amaia Vilas
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Xabier Martinez-de-Morentin
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Miren Lasaga
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Diego Alignani
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Bruno Paiva
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Ana C. Viñado
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Patxi San Martin-Uriz
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | - Juan P. Romero
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | | | | | | | - Gabriele Todisco
- Department of Molecular Medicine, University of Pavia & Unit of Precision Hematology Oncology, IRCCS S. Matteo Hospital Foundation, 27100 Pavia, Italy
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia & Unit of Precision Hematology Oncology, IRCCS S. Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Nuria Planell
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Borja Saez
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Jesper N. Tegner
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Stockholm, Sweden
- Computer, Electrical, and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Bioengineering Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| | - Felipe Prosper
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Service of Hematology and Cell Therapy, Clínica Universidad de Navarra; CCUN, Pamplona, Navarra, 31008; Spain
| | - David Gomez-Cabrero
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Stockholm, Sweden
- Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College, London WC2R 2LS, UK
- Bioengineering Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
23
|
Mesbahi Y, Trahair TN, Lock RB, Connerty P. Exploring the Metabolic Landscape of AML: From Haematopoietic Stem Cells to Myeloblasts and Leukaemic Stem Cells. Front Oncol 2022; 12:807266. [PMID: 35223487 PMCID: PMC8867093 DOI: 10.3389/fonc.2022.807266] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Despite intensive chemotherapy regimens, up to 60% of adults with acute myeloid leukaemia (AML) will relapse and eventually succumb to their disease. Recent studies suggest that leukaemic stem cells (LSCs) drive AML relapse by residing in the bone marrow niche and adapting their metabolic profile. Metabolic adaptation and LSC plasticity are novel hallmarks of leukemogenesis that provide important biological processes required for tumour initiation, progression and therapeutic responses. These findings highlight the importance of targeting metabolic pathways in leukaemia biology which might serve as the Achilles' heel for the treatment of AML relapse. In this review, we highlight the metabolic differences between normal haematopoietic cells, bulk AML cells and LSCs. Specifically, we focus on four major metabolic pathways dysregulated in AML; (i) glycolysis; (ii) mitochondrial metabolism; (iii) amino acid metabolism; and (iv) lipid metabolism. We then outline established and emerging drug interventions that exploit metabolic dependencies of leukaemic cells in the treatment of AML. The metabolic signature of AML cells alters during different biological conditions such as chemotherapy and quiescence. Therefore, targeting the metabolic vulnerabilities of these cells might selectively eradicate them and improve the overall survival of patients with AML.
Collapse
Affiliation(s)
- Yashar Mesbahi
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Patrick Connerty
- Children's Cancer Institute, Lowy Cancer Centre, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| |
Collapse
|
24
|
He M, Xu H, Liu G, Yang M, Zhang W, Li Y, Zhang H, Wang C, Zhang Y, Liu X, Xu S, Ding Y, Li Y, Gao Y, Zhang Q. Levistilide A Promotes Expansion of Human Umbilical Cord Blood Hematopoietic Stem Cells by Enhancing Antioxidant Activity. Front Pharmacol 2022; 13:806837. [PMID: 35250558 PMCID: PMC8895481 DOI: 10.3389/fphar.2022.806837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Several approaches to expand human hematopoietic stem cells (hHSCs) clinically along with retainable capability of multipotential differentiation have been reported, but only a few have advanced to evaluation in clinical trials, which limits the application of HSC-based therapy. Here we show a phthalide derivative, Levistilide A (LA), can serve as a promising molecule to expand functional human umbilical cord blood (UCB) HSCs ex vivo. An in-house screen identified LA out of nine natural products as an outstanding candidate for hHSCs expansion. Additionally, our data indicated that LA treatment not only increased the numbers of phenotype-defined HSCs, but also enhanced their colony formation ability. Xenotransplantation assays showed that LA treatment could maintain unaffected engraftment of hHSCs with multilineage differentiation capacity. Further experiments revealed that LA enhanced the antioxidant activity of hHSCs by reducing intracellular and mitochondrial reactive oxygen species (ROS) levels. The identification of LA provides a new strategy in solving the clinical issue of limited numbers of UCB HSCs.
Collapse
Affiliation(s)
- Mei He
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Guangju Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hexiao Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaolei Liu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yahui Ding
- College of Chemistry, Nankai University, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Yinghui Li
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Quan Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| |
Collapse
|
25
|
Inflammation Regulates Haematopoietic Stem Cells and Their Niche. Int J Mol Sci 2022; 23:ijms23031125. [PMID: 35163048 PMCID: PMC8835214 DOI: 10.3390/ijms23031125] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Haematopoietic stem cells (HSCs) reside in the bone marrow and are supported by the specialised microenvironment, a niche to maintain HSC quiescence. To deal with haematopoietic equilibrium disrupted during inflammation, HSCs are activated from quiescence directly and indirectly to generate more mature immune cells, especially the myeloid lineage cells. In the process of proliferation and differentiation, HSCs gradually lose their self-renewal potential. The extensive inflammation might cause HSC exhaustion/senescence and malignant transformation. Here, we summarise the current understanding of how HSC functions are maintained, damaged, or exhausted during acute, prolonged, and pathological inflammatory conditions. We also highlight the inflammation-altered HSC niche and its impact on escalating the insults on HSCs.
Collapse
|
26
|
Łacina P, Butrym A, Turlej E, Stachowicz-Suhs M, Wietrzyk J, Mazur G, Bogunia-Kubik K. BSG (CD147) Serum Level and Genetic Variants Are Associated with Overall Survival in Acute Myeloid Leukaemia. J Clin Med 2022; 11:jcm11020332. [PMID: 35054026 PMCID: PMC8779396 DOI: 10.3390/jcm11020332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Basigin (BSG, CD147) is a multifunctional protein involved in cancer cell survival, mostly by controlling lactate transport through its interaction with monocarboxylate transporters (MCTs) such as MCT1. Previous studies have found that single nucleotide polymorphisms (SNPs) in the gene coding for BSG and MCT1, as well as levels of the soluble form of BSG (sBSG), are potential biomarkers in various diseases. The goal of this study was to confirm BSG and MCT1 RNA overexpression in AML cell lines, as well as to analyse soluble BSG levels and selected BSG/MCT1 genetic variants as potential biomarkers in AML patients. We found that BSG and MCT1 were overexpressed in most AML cell lines. Soluble BSG was increased in AML patients compared to healthy controls, and correlated with various clinical parameters. High soluble BSG was associated with worse overall survival, higher bone marrow blast percentage, and higher white blood cell count. BSG SNPs rs4919859 and rs4682, as well as MCT1 SNP rs1049434, were also associated with overall survival of AML patients. In conclusion, this study confirms the importance of BSG/MCT1 in AML, and suggests that soluble BSG and BSG/MCT1 genetic variants may act as potential AML biomarkers.
Collapse
Affiliation(s)
- Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-713-709-960-236
| | - Aleksandra Butrym
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Eliza Turlej
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (E.T.); (M.S.-S.); (J.W.)
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (E.T.); (M.S.-S.); (J.W.)
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (E.T.); (M.S.-S.); (J.W.)
| | - Grzegorz Mazur
- Department of Internal, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| |
Collapse
|
27
|
Genovese E, Mirabile M, Rontauroli S, Sartini S, Fantini S, Tavernari L, Maccaferri M, Guglielmelli P, Bianchi E, Parenti S, Carretta C, Mallia S, Castellano S, Colasante C, Balliu M, Bartalucci N, Palmieri R, Ottone T, Mora B, Potenza L, Passamonti F, Voso MT, Luppi M, Vannucchi AM, Tagliafico E, Manfredini R. The Response to Oxidative Damage Correlates with Driver Mutations and Clinical Outcome in Patients with Myelofibrosis. Antioxidants (Basel) 2022; 11:antiox11010113. [PMID: 35052617 PMCID: PMC8772737 DOI: 10.3390/antiox11010113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Myelofibrosis (MF) is the Philadelphia-negative myeloproliferative neoplasm characterized by the worst prognosis and no response to conventional therapy. Driver mutations in JAK2 and CALR impact on JAK-STAT pathway activation but also on the production of reactive oxygen species (ROS). ROS play a pivotal role in inflammation-induced oxidative damage to cellular components including DNA, therefore leading to greater genomic instability and promoting cell transformation. In order to unveil the role of driver mutations in oxidative stress, we assessed ROS levels in CD34+ hematopoietic stem/progenitor cells of MF patients. Our results demonstrated that ROS production in CD34+ cells from CALR-mutated MF patients is far greater compared with patients harboring JAK2 mutation, and this leads to increased oxidative DNA damage. Moreover, CALR-mutant cells show less superoxide dismutase (SOD) antioxidant activity than JAK2-mutated ones. Here, we show that high plasma levels of total antioxidant capacity (TAC) correlate with detrimental clinical features, such as high levels of lactate dehydrogenase (LDH) and circulating CD34+ cells. Moreover, in JAK2-mutated patients, high plasma level of TAC is also associated with a poor overall survival (OS), and multivariate analysis demonstrated that high TAC classification is an independent prognostic factor allowing the identification of patients with inferior OS in both DIPSS lowest and highest categories. Altogether, our data suggest that a different capability to respond to oxidative stress can be one of the mechanisms underlying disease progression of myelofibrosis.
Collapse
Affiliation(s)
- Elena Genovese
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Margherita Mirabile
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Stefano Sartini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sebastian Fantini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Lara Tavernari
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Monica Maccaferri
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Paola Guglielmelli
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Elisa Bianchi
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sandra Parenti
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Chiara Carretta
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Selene Mallia
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
| | - Sara Castellano
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Corrado Colasante
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Manjola Balliu
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Niccolò Bartalucci
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Barbara Mora
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Francesco Passamonti
- Division of Hematology, Ospedale ASST Sette Laghi, University of Insubria, 21110 Varese, Italy; (B.M.); (F.P.)
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, 00133 Rome, Italy; (R.P.); (T.O.); (M.T.V.)
- Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00179 Rome, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Alessandro Maria Vannucchi
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, 50134 Florence, Italy; (P.G.); (M.B.); (N.B.); (A.M.V.)
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.C.); (E.T.)
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico, 41124 Modena, Italy; (C.C.); (L.P.); (M.L.)
| | - Rossella Manfredini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.G.); (M.M.); (S.R.); (S.S.); (S.F.); (L.T.); (E.B.); (S.P.); (C.C.); (S.M.)
- Correspondence:
| | | |
Collapse
|
28
|
Niederkorn M, Ishikawa C, M. Hueneman K, Bartram J, Stepanchick E, R. Bennett J, E. Culver-Cochran A, Bolanos LC, Uible E, Choi K, Wunderlich M, Perentesis JP, M. Chlon T, Filippi MD, Starczynowski DT. The deubiquitinase USP15 modulates cellular redox and is a therapeutic target in acute myeloid leukemia. Leukemia 2022; 36:438-451. [PMID: 34465865 PMCID: PMC8807387 DOI: 10.1038/s41375-021-01394-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Ubiquitin-specific peptidase 15 (USP15) is a deubiquitinating enzyme implicated in critical cellular and oncogenic processes. We report that USP15 mRNA and protein are overexpressed in human acute myeloid leukemia (AML) as compared to normal hematopoietic progenitor cells. This high expression of USP15 in AML correlates with KEAP1 protein and suppression of NRF2. Knockdown or deletion of USP15 in human and mouse AML models significantly impairs leukemic progenitor function and viability and de-represses an antioxidant response through the KEAP1-NRF2 axis. Inhibition of USP15 and subsequent activation of NRF2 leads to redox perturbations in AML cells, coincident with impaired leukemic cell function. In contrast, USP15 is dispensable for human and mouse normal hematopoietic cells in vitro and in vivo. A preclinical small-molecule inhibitor of USP15 induced the KEAP1-NRF2 axis and impaired AML cell function, suggesting that targeting USP15 catalytic function can suppress AML. Based on these findings, we report that USP15 drives AML cell function, in part, by suppressing a critical oxidative stress sensor mechanism and permitting an aberrant redox state. Furthermore, we postulate that inhibition of USP15 activity with small molecule inhibitors will selectively impair leukemic progenitor cells by re-engaging homeostatic redox responses while sparing normal hematopoiesis.
Collapse
Affiliation(s)
- Madeline Niederkorn
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Chiharu Ishikawa
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Kathleen M. Hueneman
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - James Bartram
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Emily Stepanchick
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Joshua R. Bennett
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Ashley E. Culver-Cochran
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Lyndsey C. Bolanos
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Emma Uible
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Kwangmin Choi
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Mark Wunderlich
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - John P. Perentesis
- grid.239573.90000 0000 9025 8099Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Timothy M. Chlon
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Marie-Dominique Filippi
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Daniel T. Starczynowski
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
29
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
30
|
Dong C, Zhang N, Zhang L. The Multi-Omic Prognostic Model of Oxidative Stress-Related Genes in Acute Myeloid Leukemia. Front Genet 2021; 12:722064. [PMID: 34659343 PMCID: PMC8514868 DOI: 10.3389/fgene.2021.722064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Acute myeloid leukemia (AML) is one of the most common cancers in the world, and oxidative stress is closely related to leukemia. A lot of effort has been made to improve the prognosis of AML. However, the situation remains serious. Hence, we focused on the study of prognostic genes in AML. Materials and Methods: Prognostic oxidative stress genes were screened out. The gene expression profile of AML patients was downloaded from the The Cancer Genome Atlas (TCGA) database. The oxidative stress-related model was constructed, by which the prognosis of AML patients was predicted using the two GEO GSE23143 datasets and the stability of the GSE71014 authentication model. Results: The prognostic oxidative stress genes were screened out in AML, and the prognostic genes were significantly enriched in a large number of pathways based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. There was a complex interaction between prognostic genes and transcription factors. After constructing the prediction model, the clinical predictive value of the model was discussed in a multi-omic study. We investigated the sensitivity of risk score to common chemotherapeutic agents, the influence of signaling pathways on the prognosis of AML patients, and the correlation of multiple genes with immune score and immune dysfunction. Conclusions: A highly effective prognostic risk model for AML patients was established and validated. The association of prognostic oxidative stress genes with drug sensitivity, signaling pathways, and immune infiltration was explored. The results suggested that oxidative stress genes promised to be potential prognostic biomarkers for AML, which may provide a new basis for disease management.
Collapse
Affiliation(s)
- Chao Dong
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Naijin Zhang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lijun Zhang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Shen J, Xu Y, Zhang S, Lyu S, Huo Y, Zhu Y, Tang K, Mou J, Li X, Hoyle DL, Wang M, Wang J, Li X, Wang ZZ, Cheng T. Single-cell transcriptome of early hematopoiesis guides arterial endothelial-enhanced functional T cell generation from human PSCs. SCIENCE ADVANCES 2021; 7:eabi9787. [PMID: 34516916 PMCID: PMC8442917 DOI: 10.1126/sciadv.abi9787] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/14/2021] [Indexed: 05/10/2023]
Abstract
Hematopoietic differentiation of human pluripotent stem cells (hPSCs) requires orchestration of dynamic cell and gene regulatory networks but often generates blood cells that lack natural function. Here, we performed extensive single-cell transcriptomic analyses to map fate choices and gene expression patterns during hematopoietic differentiation of hPSCs and showed that oxidative metabolism was dysregulated during in vitro directed differentiation. Applying hypoxic conditions at the stage of endothelial-to-hematopoietic transition in vitro effectively promoted the development of arterial specification programs that governed the generation of hematopoietic progenitor cells (HPCs) with functional T cell potential. Following engineered expression of the anti-CD19 chimeric antigen receptor, the T cells generated from arterial endothelium-primed HPCs inhibited tumor growth both in vitro and in vivo. Collectively, our study provides benchmark datasets as a resource to further understand the origins of human hematopoiesis and represents an advance in guiding in vitro generation of functional T cells for clinical applications.
Collapse
Affiliation(s)
- Jun Shen
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Key Laboratory of Blood Disease Cell Therapy, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Shuo Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Shuzhen Lyu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yingying Huo
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yaoyao Zhu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Department of Laboratory, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Key Laboratory of Blood Disease Cell Therapy, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Junli Mou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Key Laboratory of Blood Disease Cell Therapy, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Xinjie Li
- School of Medicine, Sun Yat-sen University, Guangzhou 510006, China
| | - Dixie L. Hoyle
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Min Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Key Laboratory of Blood Disease Cell Therapy, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China
| | - Xin Li
- School of Medicine, Sun Yat-sen University, Guangzhou 510006, China
| | - Zack Z. Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin 300020, China
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
32
|
Lu X, Han L, Busquets J, Collins M, Lodi A, Marszalek JR, Konopleva M, Tiziani S. The Combined Treatment With the FLT3-Inhibitor AC220 and the Complex I Inhibitor IACS-010759 Synergistically Depletes Wt- and FLT3-Mutated Acute Myeloid Leukemia Cells. Front Oncol 2021; 11:686765. [PMID: 34490088 PMCID: PMC8417744 DOI: 10.3389/fonc.2021.686765] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with a high mortality rate and relapse risk. Although progress on the genetic and molecular understanding of this disease has been made, the standard of care has changed minimally for the past 40 years and the five-year survival rate remains poor, warranting new treatment strategies. Here, we applied a two-step screening platform consisting of a primary cell viability screening and a secondary metabolomics-based phenotypic screening to find synergistic drug combinations to treat AML. A novel synergy between the oxidative phosphorylation inhibitor IACS-010759 and the FMS-like tyrosine kinase 3 (FLT3) inhibitor AC220 (quizartinib) was discovered in AML and then validated by ATP bioluminescence and apoptosis assays. In-depth stable isotope tracer metabolic flux analysis revealed that IACS-010759 and AC220 synergistically reduced glucose and glutamine enrichment in glycolysis and the TCA cycle, leading to impaired energy production and de novo nucleotide biosynthesis. In summary, we identified a novel drug combination, AC220 and IACS-010759, which synergistically inhibits cell growth in AML cells due to a major disruption of cell metabolism, regardless of FLT3 mutation status.
Collapse
Affiliation(s)
- Xiyuan Lu
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jonathan Busquets
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Meghan Collins
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Alessia Lodi
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Joseph R. Marszalek
- TRACTION - Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
33
|
Lévesque JP, Summers KM, Millard SM, Bisht K, Winkler IG, Pettit AR. Role of macrophages and phagocytes in orchestrating normal and pathologic hematopoietic niches. Exp Hematol 2021; 100:12-31.e1. [PMID: 34298116 DOI: 10.1016/j.exphem.2021.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
The bone marrow (BM) contains a mosaic of niches specialized in supporting different maturity stages of hematopoietic stem and progenitor cells such as hematopoietic stem cells and myeloid, lymphoid, and erythroid progenitors. Recent advances in BM imaging and conditional gene knockout mice have revealed that niches are a complex network of cells of mesenchymal, endothelial, neuronal, and hematopoietic origins, together with local physicochemical parameters. Within these complex structures, phagocytes, such as neutrophils, macrophages, and dendritic cells, all of which are of hematopoietic origin, have been found to be important in regulating several niches in the BM, including hematopoietic stem cell niches, erythropoietic niches, and niches involved in endosteal bone formation. There is also increasing evidence that these macrophages have an important role in adapting hematopoiesis, erythropoiesis, and bone formation in response to inflammatory stressors and play a key part in maintaining the integrity and function of these. Likewise, there is also accumulating evidence that subsets of monocytes, macrophages, and other phagocytes contribute to the progression and response to treatment of several lymphoid malignancies such as multiple myeloma, Hodgkin lymphoma, and non-Hodgkin lymphoma, as well as lymphoblastic leukemia, and may also play a role in myelodysplastic syndrome and myeloproliferative neoplasms associated with Noonan syndrome and aplastic anemia. In this review, the potential functions of macrophages and other phagocytes in normal and pathologic niches are discussed, as are the challenges in studying BM and other tissue-resident macrophages at the molecular level.
Collapse
Affiliation(s)
- Jean-Pierre Lévesque
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia.
| | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Susan M Millard
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Kavita Bisht
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Ingrid G Winkler
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Allison R Pettit
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
34
|
Abstract
Oxidative stress is caused by the imbalance between the generation of free radicals/reactive oxygen species (ROS) and the antioxidant defense systems, which can activate various transcription factors and affect their transcriptional pathways. Oxidative stress plays an important role in the occurrence and development of leukemia and is closely related to the treatment and prognosis of leukemia. The standard chemotherapy strategies for the pre-treatment of leukemia have many drawbacks. Hence, the usage of antioxidants and oxidants in the treatment of leukemia is being explored and has been preliminarily applied. This article reviews the research progress of oxidative stress and leukemia. In addition, the application of antioxidants treatment in leukemia has been summarized.
Collapse
|
35
|
Samimi A, Khodayar MJ, Alidadi H, Khodadi E. The Dual Role of ROS in Hematological Malignancies: Stem Cell Protection and Cancer Cell Metastasis. Stem Cell Rev Rep 2021; 16:262-275. [PMID: 31912368 DOI: 10.1007/s12015-019-09949-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Reactive oxygen species (ROS) play crucial role in hematopoiesis, regulation of differentiation, self-renewal, and the balance between quiescence and proliferation of hematopoietic stem cells (HSCs). The HSCs are a small population of undifferentiated cells that reside in the bone marrow (BM) and can undergo self-renewal by giving rise to mature cells. METHODS Relevant literature was identified through a PubMed search (2000-2019) of English-language papers using the following terms: reactive oxygen species, hematopoietic stem cell, leukemic stem cell, leukemia and chemotherapy. RESULTS HSCs are very sensitive to high levels of ROS and increased production of ROS have been attributed to HSC aging. HSC aging induced by both cell intrinsic and extrinsic factors is linked to impaired HSC self-renewal and regeneration. In addition, the elevated ROS levels might even trigger differentiation of Leukemic stem cells (LSCs) and ROS may be involved in the initiation and progression of hematological malignancies, such as leukemia. CONCLUSION Targeting genes involved in ROS in LSCs and HSCs are increasingly being used as a critical target for therapeutic interventions. Appropriate concentration of ROS may be an optimal therapeutic target for treatment of leukemia during chemotherapy, but still more studies are required to better understanding of the of ROS role in blood disorders.
Collapse
Affiliation(s)
- Azin Samimi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Legal Medicine Organization, Legal Medicine Research Center, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Alidadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elahe Khodadi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
36
|
Panina SB, Pei J, Kirienko NV. Mitochondrial metabolism as a target for acute myeloid leukemia treatment. Cancer Metab 2021; 9:17. [PMID: 33883040 PMCID: PMC8058979 DOI: 10.1186/s40170-021-00253-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemias (AML) are a group of aggressive hematologic malignancies resulting from acquired genetic mutations in hematopoietic stem cells that affect patients of all ages. Despite decades of research, standard chemotherapy still remains ineffective for some AML subtypes and is often inappropriate for older patients or those with comorbidities. Recently, a number of studies have identified unique mitochondrial alterations that lead to metabolic vulnerabilities in AML cells that may present viable treatment targets. These include mtDNA, dependency on oxidative phosphorylation, mitochondrial metabolism, and pro-survival signaling, as well as reactive oxygen species generation and mitochondrial dynamics. Moreover, some mitochondria-targeting chemotherapeutics and their combinations with other compounds have been FDA-approved for AML treatment. Here, we review recent studies that illuminate the effects of drugs and synergistic drug combinations that target diverse biomolecules and metabolic pathways related to mitochondria and their promise in experimental studies, clinical trials, and existing chemotherapeutic regimens.
Collapse
Affiliation(s)
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, USA
| | | |
Collapse
|
37
|
Oxidative Stress and ROS-Mediated Signaling in Leukemia: Novel Promising Perspectives to Eradicate Chemoresistant Cells in Myeloid Leukemia. Int J Mol Sci 2021; 22:ijms22052470. [PMID: 33671113 PMCID: PMC7957553 DOI: 10.3390/ijms22052470] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/04/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Myeloid leukemic cells are intrinsically under oxidative stress due to impaired reactive oxygen species (ROS) homeostasis, a common signature of several hematological malignancies. The present review focuses on the molecular mechanisms of aberrant ROS production in myeloid leukemia cells as well as on the redox-dependent signaling pathways involved in the leukemogenic process. Finally, the relevance of new chemotherapy options that specifically exert their pharmacological activity by altering the cellular redox imbalance will be discussed as an effective strategy to eradicate chemoresistant cells.
Collapse
|
38
|
Saulle E, Spinello I, Quaranta MT, Pasquini L, Pelosi E, Iorio E, Castelli G, Chirico M, Pisanu ME, Ottone T, Voso MT, Testa U, Labbaye C. Targeting Lactate Metabolism by Inhibiting MCT1 or MCT4 Impairs Leukemic Cell Proliferation, Induces Two Different Related Death-Pathways and Increases Chemotherapeutic Sensitivity of Acute Myeloid Leukemia Cells. Front Oncol 2021; 10:621458. [PMID: 33614502 PMCID: PMC7892602 DOI: 10.3389/fonc.2020.621458] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022] Open
Abstract
Metabolism in acute myeloid leukemia (AML) cells is dependent primarily on oxidative phosphorylation. However, in order to sustain their high proliferation rate and metabolic demand, leukemic blasts use a number of metabolic strategies, including glycolytic metabolism. Understanding whether monocarboxylate transporters MCT1 and MCT4, which remove the excess of lactate produced by cancer cells, represent new hematological targets, and whether their respective inhibitors, AR-C155858 and syrosingopine, can be useful in leukemia therapy, may reveal a novel treatment strategy for patients with AML. We analyzed MCT1 and MCT4 expression and function in hematopoietic progenitor cells from healthy cord blood, in several leukemic cell lines and in primary leukemic blasts from patients with AML, and investigated the effects of AR-C155858 and syrosingopine, used alone or in combination with arabinosylcytosine, on leukemic cell proliferation. We found an inverse correlation between MCT1 and MCT4 expression levels in leukemic cells, and showed that MCT4 overexpression is associated with poor prognosis in AML patients. We also found that AR-C155858 and syrosingopine inhibit leukemic cell proliferation by activating two different cell-death related pathways, i.e., necrosis for AR-C155858 treatment and autophagy for syrosingopine, and showed that AR-C155858 and syrosingopine exert an anti-proliferative effect, additive to chemotherapy, by enhancing leukemic cells sensitivity to chemotherapeutic agents. Altogether, our study shows that inhibition of MCT1 or MCT4 impairs leukemic cell proliferation, suggesting that targeting lactate metabolism may be a new therapeutic strategy for AML, and points to MCT4 as a potential therapeutic target in AML patients and to syrosingopine as a new anti-proliferative drug and inducer of autophagy to be used in combination with conventional chemotherapeutic agents in AML treatment.
Collapse
Affiliation(s)
- Ernestina Saulle
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Isabella Spinello
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Teresa Quaranta
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Luca Pasquini
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Egidio Iorio
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mattea Chirico
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Tiziana Ottone
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome, Italy
| | - Ugo Testa
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Catherine Labbaye
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
39
|
Nakamura-Ishizu A, Ito K, Suda T. Hematopoietic Stem Cell Metabolism during Development and Aging. Dev Cell 2021; 54:239-255. [PMID: 32693057 DOI: 10.1016/j.devcel.2020.06.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
Cellular metabolism in hematopoietic stem cells (HSCs) is an area of intense research interest, but the metabolic requirements of HSCs and their adaptations to their niches during development have remained largely unaddressed. Distinctive from other tissue stem cells, HSCs transition through multiple hematopoietic sites during development. This transition requires drastic metabolic shifts, insinuating the capacity of HSCs to meet the physiological demand of hematopoiesis. In this review, we highlight how mitochondrial metabolism determines HSC fate, and especially focus on the links between mitochondria, endoplasmic reticulum (ER), and lysosomes in HSC metabolism.
Collapse
Affiliation(s)
- Ayako Nakamura-Ishizu
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA; Department of Medicine (Hemato-Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA; Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, USA
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| |
Collapse
|
40
|
Vascular adhesion protein-1 defines a unique subpopulation of human hematopoietic stem cells and regulates their proliferation. Cell Mol Life Sci 2021; 78:7851-7872. [PMID: 34719737 PMCID: PMC8629906 DOI: 10.1007/s00018-021-03977-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 10/25/2022]
Abstract
Although the development of hematopoietic stem cells (HSC) has been studied in great detail, their heterogeneity and relationships to different cell lineages remain incompletely understood. Moreover, the role of Vascular Adhesion Protein-1 in bone marrow hematopoiesis has remained unknown. Here we show that VAP-1, an adhesin and a primary amine oxidase producing hydrogen peroxide, is expressed on a subset of human HSC and bone marrow vasculature forming a hematogenic niche. Bulk and single-cell RNAseq analyses reveal that VAP-1+ HSC represent a transcriptionally unique small subset of differentiated and proliferating HSC, while VAP-1- HSC are the most primitive HSC. VAP-1 generated hydrogen peroxide acts via the p53 signaling pathway to regulate HSC proliferation. HSC expansion and differentiation into colony-forming units are enhanced by inhibition of VAP-1. Contribution of VAP-1 to HSC proliferation was confirmed with mice deficient of VAP-1, mice expressing mutated VAP-1 and using an enzyme inhibitor. In conclusion, VAP-1 expression allows the characterization and prospective isolation of a new subset of human HSC. Since VAP-1 serves as a check point-like inhibitor in HSC differentiation, the use of VAP-1 inhibitors enables the expansion of HSC.
Collapse
|
41
|
Involvement of Oxidative Stress in Resistance to Tyrosine-Kinase Inhibitors Therapy in Chronic Myeloid Leukemia. CURRENT HEALTH SCIENCES JOURNAL 2020; 46:420-432. [PMID: 33717518 PMCID: PMC7948018 DOI: 10.12865/chsj.46.04.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/12/2020] [Indexed: 11/29/2022]
Abstract
Oxidative stress involves disruption of the cellular redox status through excessive production of reactive oxygen species or through deficiency in the cellular antioxidant capacity. It is involved in the pathogeny of multiple entities (hematological diseases, metabolic disorders, cardiovascular and renal pathology etc.), as well as in the pharmacokinetics of specific treatments for these pathologies. Chronic myeloid leukemia is a chronic myeloproliferative disease for which current standard treatment is BCR-ABL tyrosine kinase inhibitors. The innovation of this therapy has significantly improved life expectancy for patients with chronic myeloid leukemia, but in some cases, this treatment becomes ineffective, installing the resistance to tyrosine kinase inhibitors therapy. There were described two types of tyrosin kinase inhibitors resistance: primary and secondary resistance. In the present paper we proposed to evaluate the involvement of oxidative in the resistance to tyrosine kinase inhibitors therapy, in the clonal instability in chronic myeloid leukemia and in the progression of the disease to an advanced stage. We concluded that oxidative stress can play a dual role in the evolution of chronic myeloid leukemia: on the one hand it can promote genomic instability and accelerate the progression of the disease to advanced stages associated with tyrosin kinase inhibitors resistance and, on the other hand, it can contribute to leukemic cell apoptosis. It seems to be outlined a fragile balance between the pro- and anti-apoptotic effects of the reactive oxygen species, closely related to their level in the leukemic cells.
Collapse
|
42
|
Carter BZ, Mak PY, Tao W, Warmoes M, Lorenzi PL, Mak D, Ruvolo V, Tan L, Cidado J, Drew L, Andreeff M. Targeting MCL-1 dysregulates cell metabolism and leukemia-stroma interactions and resensitizes acute myeloid leukemia to BCL-2 inhibition. Haematologica 2020; 107:58-76. [PMID: 33353284 PMCID: PMC8719086 DOI: 10.3324/haematol.2020.260331] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Indexed: 12/02/2022] Open
Abstract
MCL-1 and BCL-2 are both frequently overexpressed in acute myeloid leukemia (AML) and critical for the survival of AML cells and AML stem cells. MCL-1 is a key factor in venetoclax resistance. Using genetic and pharmacological approaches, we discovered that MCL-1 regulates leukemia cell bioenergetics and carbohydrate metabolisms, including the TCA cycle, glycolysis and pentose phosphate pathway and modulates cell adhesion proteins and leukemia-stromal interactions. Inhibition of MCL-1 sensitizes to BCL-2 inhibition in AML cells and AML stem/progenitor cells, including those with intrinsic and acquired resistance to venetoclax through cooperative release of pro-apoptotic BIM, BAX, and BAK from binding to anti-apoptotic BCL- 2 proteins and inhibition of cell metabolism and key stromal microenvironmental mechanisms. The combined inhibition of MCL-1 by MCL-1 inhibitor AZD5991 or CDK9 inhibitor AZD4573 and BCL-2 by venetoclax greatly extended survival of mice bearing patient-derived xenografts established from an AML patient who acquired resistance to venetoclax/decitabine. These results demonstrate that co-targeting MCL-1 and BCL-2 improves the efficacy of and overcomes pre-existing and acquired resistance to BCL-2 inhibition. Activation of metabolomic pathways and leukemia-stroma interactions are newly discovered functions of MCL-1 in AML, which are independent from canonical regulation of apoptosis by MCL-1. Our data provide new mechanisms of synergy and a rationale for co-targeting MCL-1 and BCL-2 clinically in patients with AML and potentially other cancers.
Collapse
Affiliation(s)
- Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston.
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Wenjing Tao
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Marc Warmoes
- Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston
| | - Duncan Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston
| | | | - Lisa Drew
- Bioscience Oncology RandD, AstraZeneca, Boston
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston.
| |
Collapse
|
43
|
Yang Z, Jiang H. A chromatin perspective on metabolic and genotoxic impacts on hematopoietic stem and progenitor cells. Cell Mol Life Sci 2020; 77:4031-4047. [PMID: 32318759 PMCID: PMC7541408 DOI: 10.1007/s00018-020-03522-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/17/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
Fate determination in self-renewal and differentiation of hematopoietic stem and progenitor cells (HSCs and HPCs) is ultimately controlled by gene expression, which is profoundly influenced by the global and local chromatin state. Cellular metabolism directly influences the chromatin state through the dynamic regulation of the enzymatic activities that modify DNA and histones, but also generates genotoxic metabolites that can damage DNA and thus pose threat to the genome integrity. On the other hand, mechanisms modulating the chromatin state impact metabolism by regulating the expression and activities of key metabolic enzymes. Moreover, through regulating either DNA damage response directly or expression of genes involved in this process, chromatin modulators play active and crucial roles in guarding the genome integrity, breaching of which results in defective HSPC function. Therefore, HSPC function is regulated by the dynamic and two-way interactions between metabolism and chromatin. Here, we review recent advances that provide a chromatin perspective on the major impacts the metabolic and genotoxic factors can have on HSPC function and fate determination.
Collapse
Affiliation(s)
- Zhenhua Yang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hao Jiang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
44
|
Hypoxia Regulates Lymphoid Development of Human Hematopoietic Progenitors. Cell Rep 2020; 29:2307-2320.e6. [PMID: 31747603 DOI: 10.1016/j.celrep.2019.10.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/29/2019] [Accepted: 10/10/2019] [Indexed: 01/04/2023] Open
Abstract
Hypoxia plays a major role in the physiology of hematopoietic and immune niches. Important clues from works in mouse have paved the way to investigate the role of low O2 levels in hematopoiesis. However, whether hypoxia impacts the initial steps of human lymphopoiesis remains unexplored. Here, we show that hypoxia regulates cellular and metabolic profiles of umbilical cord blood (UCB)-derived hematopoietic progenitor cells. Hypoxia more specifically enhances in vitro lymphoid differentiation potentials of lymphoid-primed multipotent progenitors (LMPPs) and pro-T/natural killer (NK) cells and in vivo B cell potential of LMPPs. In accordance, hypoxia exacerbates the lymphoid gene expression profile through hypoxia-inducible factor (HIF)-1α (for LMPPs) and HIF-2α (for pro-T/NK). Moreover, loss of HIF-1/2α expression seriously impedes NK and B cell production from LMPPs and pro-T/NK. Our study describes how hypoxia contributes to the lymphoid development of human progenitors and reveals the implication of the HIF pathway in LMPPs and pro-T/NK-cell lymphoid identities.
Collapse
|
45
|
Progress towards improving homing and engraftment of hematopoietic stem cells for clinical transplantation. Curr Opin Hematol 2020; 26:266-272. [PMID: 31045644 DOI: 10.1097/moh.0000000000000510] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Hematopoietic cell transplantation (HCT) is a life-saving treatment for a variety of hematological and nonhematological disorders. Successful clinical outcomes after transplantation rely on adequate hematopoietic stem cell (HSC) numbers, and the homing and subsequent short-term and long-term engraftment of these cells in the bone marrow. Enhancing the homing capability of HSCs has the potential for high impact on improving HCT and patient survival. RECENT FINDINGS There are a number of ways to enhance HSC engraftment. Neutralizing negative epigenetic regulation by histone deacetylase 5 (HDAC5) increases surface CXCR4 expression and promotes human HSC homing and engraftment in immune-deficient NSG (NOD.Cg-Prkdc IL2rgt/Sz) mice. Short-term treatment of cells with glucocorticoids, pharmacological stabilization of hypoxia-inducible factor (HIF)-1α, increasing membrane lipid raft aggregation, and inhibition of dipeptidyl peptidase 4 (DPP4) facilitates HSC homing and engraftment. Added to these procedures, modulating the mitochondria permeability transition pore (MPTP) to mitigate ambient air-induced extra physiological oxygen stress/shock (EPHOSS) by hypoxic harvest and processing, or using cyclosporine A during air collection increases functional HSC numbers and improves HSC engraftment. SUMMARY A better understanding of the regulation of human HSC homing mediated by various signaling pathways will facilitate development of more efficient means to enhance HCT efficacy.
Collapse
|
46
|
Vignon C, Lachot S, Foucault A, Ravalet N, Gyan E, Picou F, Herault B, Le Nail L, Bene MC, Herault O. Reactive oxygen species levels differentiate
CD34
+
human progenitors based on
CD38
expression. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:516-521. [DOI: 10.1002/cyto.b.21948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 07/01/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Christine Vignon
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
| | - Sébastien Lachot
- Tours University Hospital Department of Biological Hematology Tours France
| | - Amélie Foucault
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
- Tours University Hospital Department of Biological Hematology Tours France
| | - Noémie Ravalet
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
- Tours University Hospital Department of Biological Hematology Tours France
| | - Emmanuel Gyan
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
- Tours University Hospital Department of Hematology and Cell Therapy Tours France
| | - Frédéric Picou
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
- Tours University Hospital Department of Biological Hematology Tours France
| | - Beatrice Herault
- French Blood Establishment (EFS) Centre‐Pays de la Loire Tours France
| | | | - Marie C. Bene
- Nantes University Hospital Hematology Biology & CIRCNA Nantes France
| | - Olivier Herault
- CNRS ERL7001 LNOx “Leukemic Niche & redox metabolism” and EA7501 GICC Tours University Tours France
- Tours University Hospital Department of Biological Hematology Tours France
| |
Collapse
|
47
|
Ramakrishnan R, Peña-Martínez P, Agarwal P, Rodriguez-Zabala M, Chapellier M, Högberg C, Eriksson M, Yudovich D, Shah M, Ehinger M, Nilsson B, Larsson J, Hagström-Andersson A, Ebert BL, Bhatia R, Järås M. CXCR4 Signaling Has a CXCL12-Independent Essential Role in Murine MLL-AF9-Driven Acute Myeloid Leukemia. Cell Rep 2020; 31:107684. [PMID: 32460032 PMCID: PMC8109054 DOI: 10.1016/j.celrep.2020.107684] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/28/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is defined by an accumulation of immature myeloid blasts in the bone marrow. To identify key dependencies of AML stem cells in vivo, here we use a CRISPR-Cas9 screen targeting cell surface genes in a syngeneic MLL-AF9 AML mouse model and show that CXCR4 is a top cell surface regulator of AML cell growth and survival. Deletion of Cxcr4 in AML cells eradicates leukemia cells in vivo without impairing their homing to the bone marrow. In contrast, the CXCR4 ligand CXCL12 is dispensable for leukemia development in recipient mice. Moreover, expression of mutated Cxcr4 variants reveals that CXCR4 signaling is essential for leukemia cells. Notably, loss of CXCR4 signaling in leukemia cells leads to oxidative stress and differentiation in vivo. Taken together, our results identify CXCR4 signaling as essential for AML stem cells by protecting them from differentiation independent of CXCL12 stimulation.
Collapse
Affiliation(s)
| | | | - Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | | | | | - Carl Högberg
- Division of Clinical Genetics, Lund University, Lund 22184, Sweden
| | - Mia Eriksson
- Division of Clinical Genetics, Lund University, Lund 22184, Sweden
| | - David Yudovich
- Division of Molecular Medicine and Gene Therapy, Lund University, Lund 22184, Sweden
| | - Mansi Shah
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Mats Ehinger
- Division of Pathology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund 22184, Sweden
| | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Lund University, Lund 22184, Sweden
| | - Jonas Larsson
- Division of Molecular Medicine and Gene Therapy, Lund University, Lund 22184, Sweden
| | | | - Benjamin L Ebert
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Marcus Järås
- Division of Clinical Genetics, Lund University, Lund 22184, Sweden.
| |
Collapse
|
48
|
Chirillo R, Aversa I, Di Vito A, Salatino A, Battaglia AM, Sacco A, Di Sanzo MA, Faniello MC, Quaresima B, Palmieri C, Biamonte F, Costanzo F. FtH-Mediated ROS Dysregulation Promotes CXCL12/CXCR4 Axis Activation and EMT-Like Trans-Differentiation in Erythroleukemia K562 Cells. Front Oncol 2020; 10:698. [PMID: 32432042 PMCID: PMC7214836 DOI: 10.3389/fonc.2020.00698] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/14/2020] [Indexed: 12/23/2022] Open
Abstract
The cell-microenvironment communication is essential for homing of hematopoietic stem cells in stromal niches. Recent evidences support the involvement of epithelial-to-mesenchymal (EMT) process in hematopoietic stem cell homeostasis as well as in leukemia cells invasiveness and migration capability. Here, we demonstrate that the alteration of iron homeostasis and the consequent increase of redox metabolism, mediated by the stable knock down of ferritin heavy chain (FtH), enhances the expression of CXCR4 in K562 erythroleukemia cells, thus promoting CXCL12-mediated motility. Indeed, addition of the CXCR4 receptor antagonist AMD3100 reverts this effect. Upon FtH knock down K562 cells also acquire an “EMT-like” phenotype, characterized by the increase of Snail, Slug and Vimentin with the parallel loss of E-cadherin. By using fibronectin as substrate, the cell adhesion assay further shows a reduction of cell adhesion capability in FtH-silenced K562 cells. Accordingly, confocal microscopy shows that adherent K562 control cells display a variety of protrusions while FtH-silenced K562 cells remain roundish. These phenomena are largely due to the reactive oxygen species (ROS)-mediated up-regulation of HIF-1α/CXCR4 axis which, in turn, promotes the activation of NF-κB and the enhancement of EMT features. These data are confirmed by treatments with either N-acetylcysteine (NAC) or AMD3100 or NF-κB inhibitor IκB-alpha which revert the FtH-silenced K562 invasive phenotype. Overall, our findings demonstrate the existence of a direct relationship among iron metabolism, redox homeostasis and EMT in the hematological malignancies. The effects of FtH dysregulation on CXCR4/CXCL12-mediated K562 cell motility extend the meaning of iron homeostasis in the leukemia cell microenvironment.
Collapse
Affiliation(s)
- Roberta Chirillo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Ilenia Aversa
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Alessandro Salatino
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Anna Martina Battaglia
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Alessandro Sacco
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Maddalena Adriana Di Sanzo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Maria Concetta Faniello
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy.,Department of Experimental and Clinical Medicine, Research Center of Biochemistry and Advanced Molecular Biology, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Barbara Quaresima
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy.,Department of Experimental and Clinical Medicine, Research Center of Biochemistry and Advanced Molecular Biology, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Francesco Costanzo
- Department of Experimental and Clinical Medicine, Research Center of Biochemistry and Advanced Molecular Biology, "Magna Græcia" University of Catanzaro, Catanzaro, Italy.,Interdepartmental Center of Services (CIS), "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
49
|
Oxidative resistance of leukemic stem cells and oxidative damage to hematopoietic stem cells under pro-oxidative therapy. Cell Death Dis 2020; 11:291. [PMID: 32341354 PMCID: PMC7184730 DOI: 10.1038/s41419-020-2488-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Leukemic stem cells (LSCs) and hematopoietic stem cells (HSCs) are both dependent on the hypoxic bone marrow (BM) microenvironment (also known as the BM niche). There is always fierce competition between the two types of cells, and the former exhibits a greater competitive advantage than the latter via multiple mechanisms. Under hypoxia, the dynamic balance between the generation and clearing of intracellular reactive oxygen species (ROS) is conducive to maintaining a quiescent state of cells. Quiescent LSCs can reside well in the BM niche, avoiding attack by chemotherapeutic agents, which is the cause of chemotherapeutic resistance and relapse in leukemia. HSCs acquire energy mainly through anaerobic glycolysis, whereas LSCs achieve energy metabolism largely through mitochondrial oxidative respiration. Mitochondria are the primary site of ROS generation. Thus, in theory, mitochondria-mediated respiration will cause an increase in ROS generation in LSCs and a higher intracellular oxidative stress level. The sensitivity of the cells to pro-oxidant drugs increases as well, which allows for the selective clearing of LSCs by pro-oxidative therapy. However, HSCs are also highly sensitive to changes in ROS levels, and the toxic effects of pro-oxidant drugs on HSCs poses a major challenge to pro-oxidative therapy in leukemia. Given the above facts, we reviewed studies on the oxidative resistance of LSCs and the oxidative damage to HSCs under pro-oxidative therapy. An in-depth investigation into the oxidative stress status and regulatory mechanisms of LSCs and HSCs in hypoxic environments will promote our understanding of the survival strategy employed by LSCs and the mechanism of the oxidative damage to HSCs in the BM niche, thus facilitating individualized treatment of leukemia patients and helping eliminate LSCs without disturbing normal hematopoietic cells.
Collapse
|
50
|
Li Y, He M, Zhang W, Yang M, Ding Y, Xu S, Gu J, Li Y, Yin J, Gao Y. Antioxidant Small Molecule Compound Chrysin Promotes the Self-Renewal of Hematopoietic Stem Cells. Front Pharmacol 2020; 11:399. [PMID: 32300303 PMCID: PMC7142222 DOI: 10.3389/fphar.2020.00399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
There is an increasing demand for the expansion of functional human hematopoietic stem cells (hHSCs) for various clinical applications. Based on our primary screening of antioxidant small molecule compounds library, a small molecule compound C2968 (chrysin) was identificated to expand cord blood CD34+ cells in vitro. Then we further verified the optimum concentration and explored its effect on hHSCs phenotype and biological function. C2968 could significantly increase the proportion and absolute number of CD34+CD38−CD49f+ and CD34+CD38−CD45RA−CD90+ cells under 2.5 μM. Furthermore, the total number of colony-forming units and the frequency of LT-HSCs in C2968-treated group were significantly higher than control, indicating the multipotency and long-term activity of hematopoietic stem and progenitor cells were sustained. Additionally, C2968 treatment could maintain transplantable HSCs that preserve balanced multilineage potential and promote rapid engraftment after transplantation in immunodeficient (NOG) mice. Mechanistically, the activity of chrysin might be mediated through multiple mechanisms namely delaying HSC differentiation, inhibiting ROS-activated apoptosis, and modulating of cyclin-dependent kinase inhibitors. Overall, chrysin showed good ex vivo expansion effect on hHSCs, which could maintain the self-renewal and multilineage differentiation potential of hHSCs. Through further research on its antioxidant mechanism, it may become a promising tool for further fundamental research and clinical umbilical cord blood transplantation of hHSCs.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yahui Ding
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Gu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jingjing Yin
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|