Abstract

Simple SummaryAML mitochondrial oxidative phosphorylation has recently been identified as a biological property that influences the response to antitumor therapy. In the present study, we propose a standardized protocol to measure mitochondrial metabolic organization in patient blasts (from the blood or bone marrow) using XFe24 or XFe96 Seahorse. Monitoring mitochondrial oxygen consumption of blasts could improve the prediction of drug response in AML patients, especially in clinical trials.Resistant acute myeloid leukemia (AML) exhibits mitochondrial energy metabolism changes compared to newly diagnosed AML. This phenotype is often observed by evaluating the mitochondrial oxygen consumption of blasts, but most of the oximetry protocols were established from leukemia cell lines without validation on primary leukemia cells. Moreover, the cultures and storage conditions of blasts freshly extracted from patient blood or bone marrow cause stress, which must be evaluated before determining oxidative phosphorylation (OXPHOS). Herein, we evaluated different conditions to measure the oxygen consumption of blasts using extracellular flow analyzers. We first determined the minimum number of blasts required to measure OXPHOS. Next, we compared the OXPHOS of blasts cultured for 3 h and 18 h after collection and found that to maintain metabolic organization for 18 h, cytokine supplementation is necessary. Cytokines are also needed when measuring OXPHOS in cryopreserved, thawed and recultured blasts. Next, the concentrations of respiratory chain inhibitors and uncoupler FCCP were established. We found that the FCCP concentration required to reach the maximal respiration of blasts varied depending on the patient sample analyzed. These protocols provided can be used in future clinical studies to evaluate OXPHOS as a biomarker and assess the efficacy of treatments targeting mitochondria.

Highlights

  • Acute myeloid leukemia (AML) is a malignant myeloid disease characterized by the loss of blast differentiation and clonal amplification in the peripheral blood and bone marrow

  • To establish a standardized protocol to assess blast oxidative phosphorylation (OXPHOS) with Seahorse XFe24 or XFe96, all experiments were performed with blasts isolated from the blood or bone marrow of AML patients (Table 1)

  • Because leukemia cells are nonadherent, they should be immobilized at the bottom of the XFe24 or XFe96 Seahorse microplates

Read more

Summary

Introduction

Acute myeloid leukemia (AML) is a malignant myeloid disease characterized by the loss of blast differentiation and clonal amplification in the peripheral blood and bone marrow. Improvements in AML diagnosis and advances in therapeutic approaches have improved the outlook for patients. Despite this considerable progress, the five-year overall survival rate for AML is still 24% [1]. European LeukemiaNet (ELN-2017) recommendations have determined the profiles of coexisting and exclusive mutations in blasts to provide prognostic information and allow for the stratification of patients into three risk categories (favorable, intermediate or adverse) [2]. The ELN classification is used to guide postremission treatment; notably, hematopoietic allogeneic transplant does not seem necessary for patients in complete first remission, whereas this therapy is strongly recommended for patients with adverse risk [3]. The evaluation of new biomarkers is needed to complete the ELN classification, and blast energy metabolism could be a potential candidate [5,6,7]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call