1
|
Fang H, Yu E, Liu C, Eapen C, Cheng C, Hu T. Metabolic landscape and rewiring in normal hematopoiesis, leukemia and aging. Semin Cancer Biol 2025:S1044-579X(25)00018-5. [PMID: 39933639 DOI: 10.1016/j.semcancer.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/06/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Recent advancements in metabolism research have demonstrated its critical roles in a lot of critical biological processes, including stemness maintenance, cell differentiation, proliferation, and function. Hematopoiesis is the fundamental cell differentiation process with the production of millions of red blood cells per second in carrying oxygen and white blood cells in fighting infection and cancers. The differentiation processes of hematopoietic stem and progenitor cells (HSPCs) are accompanied by significant metabolic reprogramming. In hematological malignancy, metabolic reprogramming is also essential to the malignant hematopoiesis processes. The metabolic rewiring is driven by distinct molecular mechanisms that meet the specific demands of different target cells. Leukemic cells, for instance, adopt unique metabolic profiles to support their heightened energy needs for survival and proliferation. Moreover, aging HSPCs exhibit altered energy consumption compared to their younger counterparts, often triggering protective mechanisms at the cellular level. In this review, we provide a comprehensive analysis of the metabolic processes involved in hematopoiesis and the metabolic rewiring that occurs under adverse conditions. In addition, we highlight current research directions and discuss the potential of targeting metabolic pathways for the management of hematological malignancies and aging.
Collapse
Affiliation(s)
- Hui Fang
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China 430071
| | - Enze Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078 Macau SAR China
| | - Chang Liu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, China 430071
| | - Christy Eapen
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912
| | - Chunming Cheng
- Stephenson Cancer Center at Oklahoma University, Oklahoma City, OK 73104.
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912.
| |
Collapse
|
2
|
Papadopoulou MT, Panagopoulou P, Paramera E, Pechlivanis A, Virgiliou C, Papakonstantinou E, Palabougiouki M, Ioannidou M, Vasileiou E, Tragiannidis A, Papakonstantinou E, Theodoridis G, Hatzipantelis E, Evangeliou A. Metabolic Fingerprint in Childhood Acute Lymphoblastic Leukemia. Diagnostics (Basel) 2024; 14:682. [PMID: 38611595 PMCID: PMC11011894 DOI: 10.3390/diagnostics14070682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/11/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is the most prevalent childhood malignancy. Despite high cure rates, several questions remain regarding predisposition, response to treatment, and prognosis of the disease. The role of intermediary metabolism in the individualized mechanistic pathways of the disease is unclear. We have hypothesized that children with any (sub)type of ALL have a distinct metabolomic fingerprint at diagnosis when compared: (i) to a control group; (ii) to children with a different (sub)type of ALL; (iii) to the end of the induction treatment. MATERIALS AND METHODS In this prospective case-control study (NCT03035344), plasma and urinary metabolites were analyzed in 34 children with ALL before the beginning (D0) and at the end of the induction treatment (D33). Their metabolic fingerprint was defined by targeted analysis of 106 metabolites and compared to that of an equal number of matched controls. Multivariate and univariate statistical analyses were performed using SIMCAP and scripts under the R programming language. RESULTS Metabolomic analysis showed distinct changes in patients with ALL compared to controls on both D0 and D33. The metabolomic fingerprint within the patient group differed significantly between common B-ALL and pre-B ALL and between D0 and D33, reflecting the effect of treatment. We have further identified the major components of this metabolic dysregulation, indicating shifts in fatty acid synthesis, transfer and oxidation, in amino acid and glycerophospholipid metabolism, and in the glutaminolysis/TCA cycle. CONCLUSIONS The disease type and time point-specific metabolic alterations observed in pediatric ALL are of particular interest as they may offer potential for the discovery of new prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Maria T. Papadopoulou
- 4th Pediatric Department, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece; (P.P.); (A.E.)
- Woman-Mother-Child Hospital, University Hospitals of Lyon, 69500 Bron, France
| | - Paraskevi Panagopoulou
- 4th Pediatric Department, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece; (P.P.); (A.E.)
| | | | - Alexandros Pechlivanis
- Department of Chemistry, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece; (A.P.)
- BIOMIC_Auth, Center for Interdisciplinary Research of the Aristotle University of Thessaloniki (CIRI), Balkan Center, 10th Km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001 Thessaloniki, Greece
| | - Christina Virgiliou
- BIOMIC_Auth, Center for Interdisciplinary Research of the Aristotle University of Thessaloniki (CIRI), Balkan Center, 10th Km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001 Thessaloniki, Greece
- Analytical Chemistry Laboratory, Department of Chemical Engineering, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | | | - Maria Palabougiouki
- Pediatric & Adolescents Hematology-Oncology Unit, 2nd Pediatric Department, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (M.P.); (M.I.); (A.T.); (E.H.)
| | - Maria Ioannidou
- Pediatric & Adolescents Hematology-Oncology Unit, 2nd Pediatric Department, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (M.P.); (M.I.); (A.T.); (E.H.)
| | - Eleni Vasileiou
- Pediatric & Adolescents Hematology-Oncology Unit, 2nd Pediatric Department, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (M.P.); (M.I.); (A.T.); (E.H.)
| | - Athanasios Tragiannidis
- Pediatric & Adolescents Hematology-Oncology Unit, 2nd Pediatric Department, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (M.P.); (M.I.); (A.T.); (E.H.)
| | | | - Georgios Theodoridis
- Department of Chemistry, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece; (A.P.)
- BIOMIC_Auth, Center for Interdisciplinary Research of the Aristotle University of Thessaloniki (CIRI), Balkan Center, 10th Km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001 Thessaloniki, Greece
| | - Emmanuel Hatzipantelis
- Pediatric & Adolescents Hematology-Oncology Unit, 2nd Pediatric Department, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (M.P.); (M.I.); (A.T.); (E.H.)
| | - Athanasios Evangeliou
- 4th Pediatric Department, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece; (P.P.); (A.E.)
- St Luke’s Hospital S.A., 55236 Pannorama, Greece
| |
Collapse
|
3
|
Pedersen S, Mikkelstrup MF, Kristensen SR, Anwardeen NR, Elrayess MA, Andreassen T. Serum NMR-Based Metabolomics Profiling Identifies Lipoprotein Subfraction Variables and Amino Acid Reshuffling in Myeloma Development and Progression. Int J Mol Sci 2023; 24:12275. [PMID: 37569650 PMCID: PMC10419104 DOI: 10.3390/ijms241512275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological cancer. It is preceded by monoclonal gammopathy of uncertain significance (MGUS)-an asymptomatic phase. It has been demonstrated that early detection increases the 5-year survival rate. However, blood-based biomarkers that enable early disease detection are lacking. Metabolomic and lipoprotein subfraction variable profiling is gaining traction to expand our understanding of disease states and, more specifically, for identifying diagnostic markers in patients with hematological cancers. This study aims to enhance our understanding of multiple myeloma (MM) and identify candidate metabolites, allowing for a more effective preventative treatment. Serum was collected from 25 healthy controls, 20 patients with MGUS, and 30 patients with MM. 1H-NMR (Nuclear Magnetic Resonance) spectroscopy was utilized to evaluate serum samples. The metabolite concentrations were examined using multivariate, univariate, and pathway analysis. Metabolic profiles of the MGUS patients revealed lower levels of alanine, lysine, leucine but higher levels of formic acid when compared to controls. However, metabolic profiling of MM patients, compared to controls, exhibited decreased levels of total Apolipoprotein-A1, HDL-4 Apolipoprotein-A1, HDL-4 Apolipoprotein-A2, HDL Free Cholesterol, HDL-3 Cholesterol and HDL-4 Cholesterol. Lastly, metabolic comparison between MGUS to MM patients primarily indicated alterations in lipoproteins levels: Total Cholesterol, HDL Cholesterol, HDL Free Cholesterol, Total Apolipoprotein-A1, HDL Apolipoprotein-A1, HDL-4 Apolipoprotein-A1 and HDL-4 Phospholipids. This study provides novel insights into the serum metabolic and lipoprotein subfraction changes in patients as they progress from a healthy state to MGUS to MM, which may allow for earlier clinical detection and treatment.
Collapse
Affiliation(s)
- Shona Pedersen
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | | | - Søren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark
| | | | - Mohamed A. Elrayess
- Biomedical Research Center (BRC), Qatar University, Doha 2713, Qatar; (N.R.A.); (M.A.E.)
| | - Trygve Andreassen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway;
- St. Olavs Hospital HF, NO-7006 Trondheim, Norway
| |
Collapse
|
4
|
Arévalo CM, Cruz-Rodriguez N, Quijano S, Fiorentino S. Plant-derived extracts and metabolic modulation in leukemia: a promising approach to overcome treatment resistance. Front Mol Biosci 2023; 10:1229760. [PMID: 37520325 PMCID: PMC10382028 DOI: 10.3389/fmolb.2023.1229760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Leukemic cells acquire complex and often multifactorial mechanisms of resistance to treatment, including various metabolic alterations. Although the use of metabolic modulators has been proposed for several decades, their use in clinical practice has not been established. Natural products, the so-called botanical drugs, are capable of regulating tumor metabolism, particularly in hematopoietic tumors, which could partly explain the biological activity attributed to them for a long time. This review addresses the most recent findings relating to metabolic reprogramming-Mainly in the glycolytic pathway and mitochondrial activity-Of leukemic cells and its role in the generation of resistance to conventional treatments, the modulation of the tumor microenvironment, and the evasion of immune response. In turn, it describes how the modulation of metabolism by plant-derived extracts can counteract resistance to chemotherapy in this tumor model and contribute to the activation of the antitumor immune system.
Collapse
Affiliation(s)
- Cindy Mayerli Arévalo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
5
|
Tsai CY, Saito T, Sarangdhar M, Abu-El-Haija M, Wen L, Lee B, Yu M, Lipata DA, Manohar M, Barakat MT, Contrepois K, Tran TH, Theoret Y, Bo N, Ding Y, Stevenson K, Ladas EJ, Silverman LB, Quadro L, Anthony TG, Jegga AG, Husain SZ. A systems approach points to a therapeutic role for retinoids in asparaginase-associated pancreatitis. Sci Transl Med 2023; 15:eabn2110. [PMID: 36921036 PMCID: PMC10205044 DOI: 10.1126/scitranslmed.abn2110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/22/2023] [Indexed: 03/17/2023]
Abstract
Among drug-induced adverse events, pancreatitis is life-threatening and results in substantial morbidity. A prototype example is the pancreatitis caused by asparaginase, a crucial drug used to treat acute lymphoblastic leukemia (ALL). Here, we used a systems approach to identify the factors affecting asparaginase-associated pancreatitis (AAP). Connectivity Map analysis of the transcriptomic data showed that asparaginase-induced gene signatures were potentially reversed by retinoids (vitamin A and its analogs). Analysis of a large electronic health record database (TriNetX) and the U.S. Federal Drug Administration Adverse Events Reporting System demonstrated a reduction in AAP risk with concomitant exposure to vitamin A. Furthermore, we performed a global metabolomic screening of plasma samples from 24 individuals with ALL who developed pancreatitis (cases) and 26 individuals with ALL who did not develop pancreatitis (controls), before and after a single exposure to asparaginase. Screening from this discovery cohort revealed that plasma carotenoids were lower in the cases than in controls. This finding was validated in a larger external cohort. A 30-day dietary recall showed that the cases received less dietary vitamin A than the controls did. In mice, asparaginase administration alone was sufficient to reduce circulating and hepatic retinol. Based on these data, we propose that circulating retinoids protect against pancreatic inflammation and that asparaginase reduces circulating retinoids. Moreover, we show that AAP is more likely to develop with reduced dietary vitamin A intake. The systems approach taken for AAP provides an impetus to examine the role of dietary vitamin A supplementation in preventing or treating AAP.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Toshie Saito
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Mayur Sarangdhar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Maisam Abu-El-Haija
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Pediatric Gastroenterology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Li Wen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Bomi Lee
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Mang Yu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Den A. Lipata
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Murli Manohar
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Monique T. Barakat
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Kévin Contrepois
- Department of Genetics, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Thai Hoa Tran
- Division of Pediatric Hematology Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montreal, QC, H3T 1C5, Canada
| | - Yves Theoret
- Département Clinique de Médecine de Laboratoire, Secteur Pharmacologie Clinique, Optilab Montréal - CHU Sainte-Justine, Montreal, H3T 1C5, Canada
| | - Na Bo
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Kristen Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Elena J. Ladas
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Institute of Human Nutrition, Columbia University, New York, NY, 10032, USA
| | - Lewis B. Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Division of Pediatric Hematology-Oncology, Boston, Children’s Hospital, Boston, MA, 02115, USA
| | - Loredana Quadro
- Department of Food Science, Rutgers Center for Lipid Research and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Tracy G. Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Sohail Z. Husain
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| |
Collapse
|
6
|
Muñiz JP, Woodhouse JP, Hughes AE, Pruitt SL, Rabin KR, Scheurer ME, Lupo PJ, Schraw JM. Residence in a Latinx enclave and end-induction minimal residual disease positivity among children with acute lymphoblastic leukemia. Pediatr Hematol Oncol 2022; 39:650-657. [PMID: 35262447 PMCID: PMC9458766 DOI: 10.1080/08880018.2022.2047850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/31/2022] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
Racial and ethnic inequities in survival persist for children with acute lymphoblastic leukemia (ALL). In the US, there are strong associations between SES, race/ethnicity, and place of residence. This is evidenced by ethnic enclaves: neighborhoods with high concentrations of ethnic residents, immigrants, and language isolation. The Latinx enclave index (LEI) can be used to investigate how residence in a Latinx enclave is associated with health outcomes. We studied the association between LEI score and minimal residual disease (MRD) in 142 pediatric ALL patients treated at Texas Children's Hospital. LEI score was associated with end-induction MRD positivity (OR per unit increase 1.63, CI 1.12-2.46). There was also a significant trend toward increased odds of MRD positivity among children living in areas with the highest enclave index scores. MRD positivity at end of induction is associated with higher incidence of relapse and lower overall survival among children with ALL; future studies are needed to elucidate the exact causes of these findings and to improve ALL outcomes among children residing within Latinx enclaves.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2047850.
Collapse
Affiliation(s)
- Joshua P Muñiz
- Department of Internal Medicine and Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - John P Woodhouse
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Amy E Hughes
- Department of Population and Data Sciences and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sandi L Pruitt
- Department of Population and Data Sciences and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Karen R Rabin
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael E Scheurer
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Philip J Lupo
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Jeremy M Schraw
- Center for Epidemiology and Population Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
Apaolaza I, San José-Enériz E, Valcarcel LV, Agirre X, Prosper F, Planes FJ. A network-based approach to integrate nutrient microenvironment in the prediction of synthetic lethality in cancer metabolism. PLoS Comput Biol 2022; 18:e1009395. [PMID: 35286311 PMCID: PMC8947600 DOI: 10.1371/journal.pcbi.1009395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/24/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
Synthetic Lethality (SL) is currently defined as a type of genetic interaction in which the loss of function of either of two genes individually has limited effect in cell viability but inactivation of both genes simultaneously leads to cell death. Given the profound genomic aberrations acquired by tumor cells, which can be systematically identified with -omics data, SL is a promising concept in cancer research. In particular, SL has received much attention in the area of cancer metabolism, due to the fact that relevant functional alterations concentrate on key metabolic pathways that promote cellular proliferation. With the extensive prior knowledge about human metabolic networks, a number of computational methods have been developed to predict SL in cancer metabolism, including the genetic Minimal Cut Sets (gMCSs) approach. A major challenge in the application of SL approaches to cancer metabolism is to systematically integrate tumor microenvironment, given that genetic interactions and nutritional availability are interconnected to support proliferation. Here, we propose a more general definition of SL for cancer metabolism that combines genetic and environmental interactions, namely loss of gene functions and absence of nutrients in the environment. We extend our gMCSs approach to determine this new family of metabolic synthetic lethal interactions. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of tumor cells, elucidating cholesterol and myo-inositol depletion as potential vulnerabilities in different malignancies. Metabolic reprogramming is one of the hallmarks of tumor cells. Synthetic lethality (SL) is a promising approach to exploit these metabolic alterations and elucidate cancer-specific genetic dependences. However, current SL approaches do not systematically consider tumor microenvironment, which is particularly important in cancer metabolism in order to generalize identified genetic dependences. In this article, we directly address this issue and propose a more general approach to SL that integrates both genetic and environmental context of tumor cells. Our definition can help to contextualize genetic dependencies in different environmental scenarios, but it could also reveal nutrient dependencies according to the genetic context. We also provide a computational pipeline to identify this new family of synthetic lethals in genome-scale metabolic networks. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of cancer cell lines, elucidating cholesterol and myo-Inositol depletion as potential vulnerabilities in different malignancies.
Collapse
Affiliation(s)
- Iñigo Apaolaza
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
| | - Edurne San José-Enériz
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Luis V. Valcarcel
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Xabier Agirre
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Felipe Prosper
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
- Clínica Universidad de Navarra, Pamplona, Spain
| | - Francisco J. Planes
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
- * E-mail:
| |
Collapse
|
8
|
Tarighat SS, Fei F, Joo EJ, Abdel-Azim H, Yang L, Geng H, Bum-Erdene K, Grice ID, von Itzstein M, Blanchard H, Heisterkamp N. Overcoming Microenvironment-Mediated Chemoprotection through Stromal Galectin-3 Inhibition in Acute Lymphoblastic Leukemia. Int J Mol Sci 2021; 22:12167. [PMID: 34830047 PMCID: PMC8624256 DOI: 10.3390/ijms222212167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
Environmentally-mediated drug resistance in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) significantly contributes to relapse. Stromal cells in the bone marrow environment protect leukemia cells by secretion of chemokines as cues for BCP-ALL migration towards, and adhesion to, stroma. Stromal cells and BCP-ALL cells communicate through stromal galectin-3. Here, we investigated the significance of stromal galectin-3 to BCP-ALL cells. We used CRISPR/Cas9 genome editing to ablate galectin-3 in stromal cells and found that galectin-3 is dispensable for steady-state BCP-ALL proliferation and viability. However, efficient leukemia migration and adhesion to stromal cells are significantly dependent on stromal galectin-3. Importantly, the loss of stromal galectin-3 production sensitized BCP-ALL cells to conventional chemotherapy. We therefore tested novel carbohydrate-based small molecule compounds (Cpd14 and Cpd17) with high specificity for galectin-3. Consistent with results obtained using galectin-3-knockout stromal cells, treatment of stromal-BCP-ALL co-cultures inhibited BCP-ALL migration and adhesion. Moreover, these compounds induced anti-leukemic responses in BCP-ALL cells, including a dose-dependent reduction of viability and proliferation, the induction of apoptosis and, importantly, the inhibition of drug resistance. Collectively, these findings indicate galectin-3 regulates BCP-ALL cell responses to chemotherapy through the interactions between leukemia cells and the stroma, and show that a combination of galectin-3 inhibition with conventional drugs can sensitize the leukemia cells to chemotherapy.
Collapse
Affiliation(s)
- Somayeh S. Tarighat
- Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (S.S.T.); (F.F.); (E.J.J.); (H.A.-A.)
| | - Fei Fei
- Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (S.S.T.); (F.F.); (E.J.J.); (H.A.-A.)
| | - Eun Ji Joo
- Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (S.S.T.); (F.F.); (E.J.J.); (H.A.-A.)
- Department of Systems Biology, Beckman Research Institute, City of Hope, Monrovia, CA 91016, USA;
| | - Hisham Abdel-Azim
- Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (S.S.T.); (F.F.); (E.J.J.); (H.A.-A.)
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Monrovia, CA 91016, USA;
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA;
| | - Khuchtumur Bum-Erdene
- Institute for Glycomics, Griffith University, Gold Coast, Southport, QLD 4222, Australia; (K.B.-E.); (I.D.G.); (M.v.I.); (H.B.)
| | - I. Darren Grice
- Institute for Glycomics, Griffith University, Gold Coast, Southport, QLD 4222, Australia; (K.B.-E.); (I.D.G.); (M.v.I.); (H.B.)
- School of Medical Science, Griffith University, Gold Coast, Southport, QLD 4222, Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, Southport, QLD 4222, Australia; (K.B.-E.); (I.D.G.); (M.v.I.); (H.B.)
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast, Southport, QLD 4222, Australia; (K.B.-E.); (I.D.G.); (M.v.I.); (H.B.)
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Nora Heisterkamp
- Division of Hematology/Oncology and Bone Marrow Transplant, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (S.S.T.); (F.F.); (E.J.J.); (H.A.-A.)
- Department of Systems Biology, Beckman Research Institute, City of Hope, Monrovia, CA 91016, USA;
| |
Collapse
|
9
|
Comparison of the blood, bone marrow, and cerebrospinal fluid metabolomes in children with b-cell acute lymphoblastic leukemia. Sci Rep 2021; 11:19613. [PMID: 34608220 PMCID: PMC8490393 DOI: 10.1038/s41598-021-99147-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Metabolomics may shed light on treatment response in childhood acute lymphoblastic leukemia (ALL), however, most assessments have analyzed bone marrow or cerebrospinal fluid (CSF), which are not collected during all phases of therapy. Blood is collected frequently and with fewer risks, but it is unclear whether findings from marrow or CSF biomarker studies may translate. We profiled end-induction plasma, marrow, and CSF from N = 10 children with B-ALL using liquid chromatography-mass spectrometry. We estimated correlations between plasma and marrow/CSF metabolite abundances detected in ≥ 3 patients using Spearman rank correlation coefficients (rs). Most marrow metabolites were detected in plasma (N = 661; 81%), and we observed moderate-to-strong correlations (median rs 0.62, interquartile range [IQR] 0.29–0.83). We detected 328 CSF metabolites in plasma (90%); plasma-CSF correlations were weaker (median rs 0.37, IQR 0.07–0.70). We observed plasma-marrow correlations for metabolites in pathways associated with end-induction residual disease (pyruvate, asparagine) and plasma-CSF correlations for a biomarker of fatigue (gamma-glutamylglutamine). There is considerable overlap between the plasma, marrow, and CSF metabolomes, and we observed strong correlations for biomarkers of clinically relevant phenotypes. Plasma may be suitable for biomarker studies in B-ALL.
Collapse
|
10
|
Saito T, Wei Y, Wen L, Srinivasan C, Wolthers BO, Tsai CY, Harris MH, Stevenson K, Byersdorfer C, Oparaji JA, Fernandez C, Mukherjee A, Abu-El-Haija M, Agnihotri S, Schmiegelow K, Showalter MR, Fogle PW, McCulloch S, Contrepois K, Silverman LB, Ding Y, Husain SZ. Impact of acute lymphoblastic leukemia induction therapy: findings from metabolomics on non-fasted plasma samples from a biorepository. Metabolomics 2021; 17:64. [PMID: 34175981 PMCID: PMC11446541 DOI: 10.1007/s11306-021-01814-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/15/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is among the most common cancers in children. With improvements in combination chemotherapy regimens, the overall survival has increased to over 90%. However, the current challenge is to mitigate adverse events resulting from the complex therapy. Several chemotherapies intercept cancer metabolism, but little is known about their collective role in altering host metabolism. OBJECTIVES We profiled the metabolomic changes in plasma of ALL patients initial- and post- induction therapy. METHODS We exploited a biorepository of non-fasted plasma samples derived from the Dana Farber Cancer Institute ALL Consortium; these samples were obtained from 50 ALL patients initial- and post-induction therapy. Plasma metabolites and complex lipids were analyzed by high resolution tandem mass spectrometry and differential mobility tandem mass spectrometry. Data were analyzed using a covariate-adjusted regression model with multiplicity adjustment. Pathway enrichment analysis and co-expression network analysis were performed to identify unique clusters of molecules. RESULTS More than 1200 metabolites and complex lipids were identified in the total of global metabolomics and lipidomics platforms. Over 20% of those molecules were significantly altered. In the pathway enrichment analysis, lipids, particularly phosphatidylethanolamines (PEs), were identified. Network analysis indicated that the bioactive fatty acids, docosahexaenoic acid (DHA)-containing (22:6) triacylglycerols (TAGs), were decreased in the post-induction therapy. CONCLUSION Metabolomic profiling in ALL patients revealed a large number of alterations following induction chemotherapy. In particular, lipid metabolism was substantially altered. The changes in metabolites and complex lipids following induction therapy could provide insight into the adverse events experienced by ALL patients.
Collapse
Affiliation(s)
- Toshie Saito
- Department of Pediatrics, Stanford University, 750 Welch Road, Palo Alto, CA, 94304, USA
| | - Yue Wei
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Li Wen
- Department of Gastroenterology and Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chaitanya Srinivasan
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin O Wolthers
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Cheng-Yu Tsai
- Department of Pediatrics, Stanford University, 750 Welch Road, Palo Alto, CA, 94304, USA
| | - Marian H Harris
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Kristen Stevenson
- Department of Data Sciences at Dana-Farber Cancer Institute, Boston, MA, USA
| | - Craig Byersdorfer
- Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular Therapies, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Christian Fernandez
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amitava Mukherjee
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maisam Abu-El-Haija
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sameer Agnihotri
- School of Medicine, Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Kevin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ying Ding
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sohail Z Husain
- Department of Pediatrics, Stanford University, 750 Welch Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
11
|
Lupo PJ, Petrick LM, Hoang TT, Janitz AE, Marcotte EL, Schraw JM, Arora M, Scheurer ME. Using primary teeth and archived dried spots for exposomic studies in children: Exploring new paths in the environmental epidemiology of pediatric cancer. Bioessays 2021; 43:e2100030. [PMID: 34106479 DOI: 10.1002/bies.202100030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022]
Abstract
It is estimated that 300,000 children 0-14 years of age are diagnosed with cancer worldwide each year. While the absolute risk of cancer in children is low, it is the leading cause of death due to disease in children in high-income countries. In spite of this, the etiologies of pediatric cancer are largely unknown. Environmental exposures have long been thought to play an etiologic role. However, to date, there are few well-established environmental risk factors for pediatric malignancies, likely due to technical barriers in collecting biological samples prospectively in pediatric populations for direct measurements. In this review, we propose the use of novel or underutilized biospecimens (dried blood spots and teeth) and molecular approaches for exposure assessment (epigenetics, metabolomics, and somatic mutational profiles). Future epidemiologic studies of pediatric cancer should incorporate novel exposure assessment methodologies, data on molecular features of tumors, and a more complete assessment of gene-environment interactions.
Collapse
Affiliation(s)
- Philip J Lupo
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Lauren M Petrick
- The Senator Frank R. Lautenberg Environmental Health Science Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thanh T Hoang
- Epidemiology Branch, National Institutes of Health, Department of Health and Human Services, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Amanda E Janitz
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Erin L Marcotte
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeremy M Schraw
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Manish Arora
- The Senator Frank R. Lautenberg Environmental Health Science Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael E Scheurer
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Brown AL, Sok P, Taylor O, Woodhouse JP, Bernhardt MB, Raghubar KP, Kahalley LS, Lupo PJ, Hockenberry MJ, Scheurer ME. Cerebrospinal Fluid Metabolomic Profiles Associated With Fatigue During Treatment for Pediatric Acute Lymphoblastic Leukemia. J Pain Symptom Manage 2021; 61:464-473. [PMID: 32889041 PMCID: PMC7914130 DOI: 10.1016/j.jpainsymman.2020.08.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/22/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
Abstract
CONTEXT Cancer-related fatigue (CRF) is one of the most distressing and persistent symptoms reported during pediatric acute lymphoblastic leukemia (ALL) therapy; however, information on the pathways underlying CRF severity is limited. OBJECTIVES We conducted global metabolomics profiling of cerebrospinal fluid (CSF) samples to provide insight into the underlying mechanisms of CRF. METHODS Fatigue in pediatric ALL patients (2012-2017) was assessed during postinduction therapy approximately six months after diagnosis. Postinduction CSF was collected from 171 participants, comprising discovery (n = 86) and replication (n = 85) cohorts. We also conducted secondary validation using diagnostic CSF from 48 replication cohort participants. CSF metabolomic profiling was performed using gas chromatography-mass spectrometry (MS) and liquid chromatography-MS/MS. Kendall's rank correlation was used to evaluate associations between metabolite abundance and CRF. False discovery rate was used to account for multiple comparisons. RESULTS Participants were 56% males and 59% Hispanic with a mean age at diagnosis of 8.5 years. A total of 274 CSF-derived metabolites were common to the discovery and replication cohorts. Eight metabolites were significantly associated with fatigue in the discovery cohort (P < 0.05), of which three were significant in the replication cohort, including false discovery rate-corrected associations with gamma-glutamylglutamine (Pcombined = 6.2E-6) and asparagine (Pcombined = 3.5E-4). Notably, the abundance of gamma-glutamylglutamine in diagnostic CSF samples was also significantly associated with fatigue (P = 0.0062). CONCLUSION The metabolites identified in our assessment have been implicated in neurotransmitter transportation and glutathione recycling, suggesting that glutamatergic pathways or oxidative stress may contribute to ALL-associated CRF. This information could inform targeted therapies for reducing CRF in at-risk individuals.
Collapse
Affiliation(s)
- Austin L Brown
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.
| | - Pagna Sok
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Olga Taylor
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - John P Woodhouse
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - M Brooke Bernhardt
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | - Lisa S Kahalley
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Philip J Lupo
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | - Michael E Scheurer
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
13
|
Zhang X, Wang J, Zhuang J, Liu C, Gao C, Li H, Ma X, Li J, Sun C. A Novel Glycolysis-Related Four-mRNA Signature for Predicting the Survival of Patients With Breast Cancer. Front Genet 2021; 12:606937. [PMID: 33584825 PMCID: PMC7876610 DOI: 10.3389/fgene.2021.606937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/04/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Glycolysis is critical in the occurrence and development of tumors. Owing to the biological and clinical heterogeneity of patients with BRCA, the traditional predictive classification system is far from satisfactory. Survival and prognosis biomarkers related to glycolysis have broad application prospects for assessing the risk of patients and guiding their individualized treatment. Methods: The mRNA expression profiles and clinical information of patients with BRCA were obtained from TCGA database, and glycolysis-related genes were obtained by GSEA. Patients with BRCA were randomly divided into the training cohort and testing cohort. Univariate and multivariate Cox analyses were used to establish and validate a new mRNA signature for predicting the prognosis of patients with BRCA. Results: We established a four-gene breast cancer prediction signature that included PGK1, SDHC, PFKL, and NUP43. The patients with BRCA in the training cohort and testing cohort were divided into high-risk and low-risk groups based on the signature. The AUC values were 0.74 (training cohort), 0.806 (testing cohort) and 0.769 (entire cohort), thereby showing that the prediction performance of the signature is acceptable. Additionally, Cox regression analysis revealed that four-gene signature could independently predict the prognosis of BRCA patients without being affected by clinical factors. Conclusion: We constructed a four-gene signature to predict the prognosis of patients with BRCA. This signature will aid in the early diagnosis and personalized treatment of breast cancer, but the specific associated biological mechanism requires further study.
Collapse
Affiliation(s)
- Xiaolu Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
14
|
Aasebø E, Berven FS, Hovland R, Døskeland SO, Bruserud Ø, Selheim F, Hernandez-Valladares M. The Progression of Acute Myeloid Leukemia from First Diagnosis to Chemoresistant Relapse: A Comparison of Proteomic and Phosphoproteomic Profiles. Cancers (Basel) 2020; 12:cancers12061466. [PMID: 32512867 PMCID: PMC7352627 DOI: 10.3390/cancers12061466] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy. Nearly 50% of the patients who receive the most intensive treatment develop chemoresistant leukemia relapse. Although the leukemogenic events leading to relapse seem to differ between patients (i.e., regrowth from a clone detected at first diagnosis, progression from the original leukemic or preleukemic stem cells), a common characteristic of relapsed AML is increased chemoresistance. The aim of the present study was to investigate at the proteomic level whether leukemic cells from relapsed patients present overlapping molecular mechanisms that contribute to this chemoresistance. We used liquid chromatography–tandem mass spectrometry (LC–MS/MS) to compare the proteomic and phosphoproteomic profiles of AML cells derived from seven patients at the time of first diagnosis and at first relapse. At the time of first relapse, AML cells were characterized by increased levels of proteins important for various mitochondrial functions, such as mitochondrial ribosomal subunit proteins (MRPL21, MRPS37) and proteins for RNA processing (DHX37, RNA helicase; RPP40, ribonuclease P component), DNA repair (ERCC3, DNA repair factor IIH helicase; GTF2F1, general transcription factor), and cyclin-dependent kinase (CDK) activity. The levels of several cytoskeletal proteins (MYH14/MYL6/MYL12A, myosin chains; VCL, vinculin) as well as of proteins involved in vesicular trafficking/secretion and cell adhesion (ITGAX, integrin alpha-X; CD36, platelet glycoprotein 4; SLC2A3, solute carrier family 2) were decreased in relapsed cells. Our study introduces new targetable proteins that might direct therapeutic strategies to decrease chemoresistance in relapsed AML.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
| | - Frode S. Berven
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Randi Hovland
- Department for Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway;
- Department of Biological Sciences, University of Bergen, 5006 Bergen, Norway
| | | | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
| | - Frode Selheim
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Maria Hernandez-Valladares
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (Ø.B.)
- The Department of Biomedicine, The Proteomics Unit at the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- Correspondence: ; Tel.: +47-5558-6368
| |
Collapse
|
15
|
Sbirkov Y, Burnusuzov H, Sarafian V. Metabolic reprogramming in childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 2020; 67:e28255. [PMID: 32293782 DOI: 10.1002/pbc.28255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022]
Abstract
The first observations of altered metabolism in malignant cells were made nearly 100 years ago and therapeutic strategies targeting cell metabolism have been in clinical use for several decades. In this review, we summarize our current understanding of cell metabolism dysregulation in childhood acute lymphoblastic leukemia (cALL). Reprogramming of cellular bioenergetic processes can be expected in the three distinct stages of cALL: at diagnosis, during standard chemotherapy, and in cases of relapse. Upregulation of glycolysis, dependency on anaplerotic energy sources, and activation of the electron transport chain have all been observed in cALL. While the current treatment strategies are tackling some of these aberrations, cALL cells are likely to be able to rewire their metabolism in order to escape therapy, which may contribute to a refractory disease and relapse. Finally, here we focus on novel therapeutic approaches emerging from our evolving understanding of the alterations of different metabolic networks in lymphoblasts.
Collapse
Affiliation(s)
- Yordan Sbirkov
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University- Plovdiv, Plovdiv, Bulgaria
| | - Hasan Burnusuzov
- Research Institute at Medical University- Plovdiv, Plovdiv, Bulgaria.,Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University- Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
16
|
El-Ghammaz AMS, Azzazi MO, Mostafa N, Hegab HM, Mahmoud AA. Prognostic significance of serum progranulin level in de novo adult acute lymphoblastic leukemia patients. Clin Exp Med 2020; 20:269-276. [PMID: 32006270 DOI: 10.1007/s10238-020-00610-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/27/2020] [Indexed: 01/14/2023]
Abstract
Increased expression of progranulin (PGRN) has been reported in some hematological cancers, but limited information regarding its significance in acute lymphoblastic leukemia (ALL) is available. This study involved 60 subjects (40 de novo adult ALL patients and 20 controls). Serum PGRN level was measured by enzyme-linked immunosorbent assay and was correlated with patient outcome. Serum PGRN level was significantly higher in patients than controls. Serum PGRN level did not correlate with age, total leukocytic count, hemoglobin, platelets, absolute blast count in peripheral blood, lactate dehydrogenase, percent of blasts in bone marrow, gender, comorbidities, the presence of central nervous system infiltration, ALL phenotype, cytogenetics and risk of the disease. High serum PGRN level was not associated with inferior overall survival (OS) on univariate analysis. Regarding cumulative incidence of relapse (CIR) and disease-free survival (DFS), high PGRN level was associated with poor results on univariate analysis. Moreover, it tended to be independent risk factor on multivariate analysis for CIR but was not an independent predictor of inferior DFS. Serum PGRN level is significantly elevated in de novo adult ALL patients and may be used as a predictor of increased relapse risk.
Collapse
Affiliation(s)
- Amro M S El-Ghammaz
- Clinical Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Mohamed O Azzazi
- Clinical Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nevine Mostafa
- Clinical Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hany M Hegab
- Clinical Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amir A Mahmoud
- Clinical Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|