scispace - formally typeset
Search or ask a question

How is daunorubine uptaked in aml cells? 


Best insight from top research papers

Daunorubicin (DNR) uptake in acute myeloid leukemia (AML) cells is a complex process influenced by various factors. Studies have shown that DNR uptake is similar across different AML cell lines, with variations in metabolism and reductase activity. In AML patients, DNR uptake is higher in untreated individuals compared to those previously treated, correlating with achieving complete remission. The intracellular distribution of DNR in AML cells differs based on the differentiation phenotype, with CD34+ cells showing perinuclear sequestration. Flow cytometry and high-performance liquid chromatography have been utilized to measure DNR uptake in AML cells, with correlations observed between drug uptake and clinical responses. These findings highlight the importance of understanding the mechanisms of DNR uptake in AML cells to optimize treatment outcomes.

Answers from top 5 papers

More filters
Papers (5)Insight
Daunorubicin uptake in AML cells involves evaluating critical parameters for isolation and extraction methods, with cellularity affecting drug recovery, leading to variable intracellular levels independent of plasma kinetics.
Daunorubicin uptake in AML cells is similar across human myeloid leukemia and erythroleukemia cell lines, with variable metabolism to daunorubicinol and reductase activity levels.
Daunorubicin is taken up differently in AML cells based on their differentiation status; immature CD34+ cells sequester it in perinuclear vesicles, while mature cells show nuclear and cytoplasmic distribution.
Daunorubicin uptake in AML cells is measured by flow cytometry, showing correlations with cellular concentrations determined by HPLC, with higher uptake in complete remission patients.
Daunorubicin uptake in AML cells correlates with treatment response. Untreated patients show higher uptake than treated ones. CsA enhances uptake, not solely dependent on mdr1 expression.

Related Questions

How to treat acute myeloid leukemia in france?5 answersIn France, the treatment of acute myeloid leukemia (AML) involves various regimens tailored to different patient profiles. Anthracycline-free regimens like TA, FLAG, and CLARA are utilized for patients ineligible for standard therapy due to cardiac comorbidities or prior anthracycline exposure. Additionally, gemtuzumab ozogamicin (GO) has shown promise in treating newly diagnosed or relapsed/refractory AML patients, with a 70% overall response rate and viable outcomes for allogeneic hematopoietic stem cell transplantation (HSCT). In elderly AML patients, low-intensity therapies like azacitidine have demonstrated efficacy, with a role in selected cases for improved overall survival compared to best supportive care. Furthermore, CNS involvement at pediatric AML diagnosis does not independently impact prognosis, with triple intrathecal chemotherapy being a recommended treatment approach. These diverse strategies highlight the importance of personalized and evolving treatment approaches for AML patients in France.
What are the laboratory tests conducted for AML?4 answersLaboratory tests for Acute Myeloid Leukemia (AML) include various methods to detect Minimal Residual Disease (MRD) and genetic mutations. Techniques like FISH, RT-PCR, ddPCR, multiparametric flow cytometry, and Next-Generation Sequencing (NGS) are utilized for MRD detection in AML patients. Additionally, genetic mutations like FLT3-ITD and FLT3-TKD are significant in AML prognosis, with FLT3 mutations occurring in a substantial percentage of AML cases. Raman spectroscopy is also emerging as a potential tool for AML diagnostics, particularly in identifying mutations like IDH1/2, offering insights into cellular changes at a molecular level. Furthermore, expanding gene panels beyond the traditional CEPBA, FLT3, and NPM1 can provide a more comprehensive risk profile in AML cases, aiding in better prognostication and treatment decisions.
Aml and dlbcl treatments with aptamers5 answersAptamer technology has shown potential for precision therapy in the treatment of lymphoma, including anaplastic large cell lymphoma (ALCL) and acute myeloid leukemia (AML). In the case of ALCL, aptamer-nanomedicine formulations have been developed for targeted chemotherapy and gene therapy. These formulations specifically bind to ALCL cells, delivering chemotherapeutic drugs and siRNA to silence the ALK oncogene, resulting in effective killing of lymphoma cells with minimal toxicity to off-target cells. In the case of AML, combination therapies using aptamers have been explored to target apoptosis. Screening studies have identified potential drug pairings that show significant synergism in AML cell lines, providing insights into novel combination treatments for AML. Overall, aptamer technology holds promise for the development of targeted and effective treatments for lymphoma, including AML and ALCL.
What is cell isolation in AML?5 answersCell isolation in acute myeloid leukemia (AML) refers to the process of separating specific cells from a sample potentially containing various cells in order to study their characteristics and functions. Several methods have been developed for isolating cells in AML, including the use of extracellular vesicles (EVs) and single cell genomics. EV trafficking plays a crucial role in shaping the bone marrow microenvironment into a leukemia-permissive niche, and different isolation methods have been tested for their efficacy and compatibility with functional assays. Single cell genomics has allowed for the characterization of cellular heterogeneity in AML, providing opportunities for studying disease biology and clinical management. These methods have enabled researchers to identify and track genetically distinct subclones in individual AML patients, which can have different regulatory phenotypes, drug sensitivities, and growth behaviors. Overall, cell isolation techniques in AML are essential for understanding the complexity of the disease and developing targeted treatments.
AML and CML?5 answersAML and CML are both types of leukemia. AML, or Acute Myeloid Leukemia, is a blood cell disorder that arises from gene mutations involved in hematopoiesis. It can develop de novo or progress from myeloproliferative disorders or myelodysplastic syndrome. CML, or Chronic Myeloid Leukemia, is characterized by uncontrolled proliferation of myeloid progenitors and is associated with the cytochrome P450 enzyme. LSCs, or leukemic stem cells, play a role in both AML and CML and can be refractory to standard therapies. The prevalence of CYP1A1*2C polymorphism in AML and CML patients was studied, but no significant differences were found in the genotypes. A CNN combined with transfer learning was used to classify and diagnose AML, ALL, and CML based on bone marrow cell microscopy images, resulting in improved accuracy compared to manual microscopy. Overall, understanding the characteristics and markers of LSCs in AML and CML is important for developing targeted therapies.
Wich Biomarker used for AML?4 answersBiomarkers used for acute myeloid leukemia (AML) include measurable residual disease (MRD), Schlafen11 (SLFN11), proteomic biomarker panels, and ITGAX. MRD is a strong prognostic marker in AML, with MRD-negative patients having lower relapse rates and better survival. SLFN11 has been associated with response to chemotherapy agents in AML, and low SLFN11 levels predict poor response to standard chemotherapy agents. Proteomic biomarker panels have the potential to improve the prognostic classification of AML patients. ITGAX has been identified as a potential prognostic factor and therapeutic target for AML. These biomarkers can provide valuable information for assessing treatment response, predicting outcomes, and guiding therapeutic decisions in AML.

See what other people are reading

Which cell line express less vimentin?
5 answers
The AGS gastric cancer cell line expresses less vimentin compared to other cell lines. Research involving AGS cells treated with metformin and vimentin-specific siRNA demonstrated a significant decrease in vimentin expression, cell motility, migration, and invasion abilities. In contrast, studies on breast cancer cell lines MCF-7 and MDA-MB-231 showed that vimentin expression led to changes in cell shape, increased cell migration, and downregulation of genes maintaining cell-cell junctions. Additionally, erythroid cells differentiated from adult peripheral blood stem cells, iPSC, and ESC exhibited persistent vimentin expression, potentially impeding enucleation in iPSC and ESC-derived erythroid cells. Therefore, among the cell lines studied, AGS cells express less vimentin compared to MCF-7, MDA-MB-231, and erythroid cells derived from stem cells.
What is the disutility value of cytokine release syndrome in multiple myeloma patients?
5 answers
The disutility value of cytokine release syndrome (CRS) in multiple myeloma patients is significant, impacting patient survival and treatment outcomes. Studies have shown that CRS incidence is high in patients receiving chimeric antigen receptor T (CAR-T) cell therapy for multiple myeloma, with severe CRS associated with shorter progression-free survival and overall survival compared to mild CRS. Effective management of CRS, including early detection and intervention with tocilizumab and dexamethasone, is crucial to prevent life-threatening complications and improve patient outcomes during CAR-T cell therapy for multiple myeloma. Teclistamab, a bispecific antibody used in multiple myeloma treatment, has also been associated with a high incidence of CRS, further emphasizing the importance of managing this adverse event in improving patient quality of life and treatment efficacy.
How does ATRA treat APML?
4 answers
ATRA (all-trans retinoic acid) is a crucial component in the treatment of Acute Promyelocytic Leukemia (APML). ATRA, when combined with chemotherapy or arsenic trioxide, has shown significant efficacy in inducing differentiation and improving survival rates in APML patients. Studies have demonstrated that ATRA, in combination with anthracycline-based chemotherapy, leads to high complete remission rates and reduced relapse incidences in newly diagnosed APL patients. Additionally, ATRA combined with arsenic trioxide has shown comparable effectiveness to ATRA plus chemotherapy in non-high-risk APL cases. The mechanism involves ATRA-induced differentiation in AML cells, with signaling pathways like STAT3, Akt, and JNK playing crucial roles in this process. Overall, ATRA plays a pivotal role in the targeted therapy of APML, leading to improved outcomes and survival rates in affected individuals.
How to know when to refer a acute lymphocytic leukemia to palliative care?
4 answers
To determine when to refer a patient with acute lymphocytic leukemia (ALL) to palliative care, it is crucial to consider standardized criteria for early palliative care integration. Patients with ALL often experience a high symptom burden, including pain, which can be distressing and predictable during induction chemotherapy. Factors such as younger age, worse performance status, ALL diagnosis, and time from chemotherapy onset can indicate a need for palliative care referral. However, the integration of palliative care in hematologic malignancies like ALL is often delayed due to various factors, including curative goals and limited access to palliative care services. Implementing strategies to increase early palliative care utilization, such as close collaboration between hematologists and palliative care teams, can improve symptom management and quality of life for patients with ALL.
What is the concentration of doxorubicin for spheroid in fluorescence microscopy?
5 answers
The concentration of doxorubicin (DOX) used for spheroids in fluorescence microscopy varies based on the study. In one study, a concentration of 25 μM of DOX was utilized for fluorescence microscopy analysis. Another study employed a concentration of 400 nM of DOX for examining the effects on multicellular spheroids (MCS). Additionally, a different research paper mentioned the use of a higher concentration of 10 μM of DOX for confocal laser scanning microscopy evaluation in MCS. These varying concentrations of DOX were crucial for assessing its distribution within spheroids and its impact on cell viability, highlighting the importance of dosage selection in fluorescence microscopy studies on spheroids.
How does global harmonization benefit manufacturers and researchers involved in developing tissue-engineered products?
5 answers
Global harmonization in the development of tissue-engineered products benefits manufacturers and researchers by facilitating standardized procedures, enhancing comparability, and streamlining regulatory processes. By aligning definitions, regulations, and jurisdictional assignments internationally, manufacturers can operate more efficiently across borders, reducing redundancies and costs. Standardization efforts also extend to critical quality assays, aiming to harmonize identity, purity, safety, and potency assessments for cell therapies. Moreover, harmonized culture conditions for mesenchymal stromal cells (MSCs) have shown reproducible results across different laboratories, emphasizing the importance of standardized protocols in research. Overall, global harmonization fosters collaboration, accelerates innovation, and ensures the safety and efficacy of tissue-engineered products on a worldwide scale.
How do mesenchymal stem cells influence the differentiation of CD4 lymphocytes?
5 answers
Mesenchymal stem cells (MSCs) play a crucial role in influencing the differentiation of CD4 lymphocytes. Studies have shown that MSCs, particularly those from the lymph nodes, produce MCP-1, promoting T-cell proliferation and differentiation. Additionally, MSCs from the amniotic membrane of the placenta impact the commitment of CD4+ T cells towards memory subsets by modulating key transcription factors and signaling pathways. Furthermore, osteogenic differentiation of MSCs affects the migration and differentiation of CD4+ T cells, with increased CD4+ T cell migration and weakened regulatory T cell differentiation observed. Autophagy in MSCs also plays a role in mediating CD4+ T cell migration and differentiation through the secretion of CXCL8 and TGF-β1. These findings provide valuable insights into the immunoregulatory functions of MSCs in modulating CD4 lymphocyte differentiation.
Who iptp guidelines?
5 answers
The guidelines for idiopathic thickened pituitary stalk (iTPS) and/or central diabetes insipidus (iCDI) in children and young people were developed by a joint society (BSPED/CCLG) multidisciplinary national endeavor following NICE methodology and overseen by RCPCH. These guidelines aimed to address the investigation and management of iTPS and iCDI cases where the etiology is not initially apparent, focusing on age-appropriate decision-making management to enhance national best practices and care quality for affected individuals. The development process involved a systematic literature search, expert opinion drafting, and a formal Delphi consensus process due to the lack of high-quality evidence for many clinical questions. The resulting decision-making flowchart accompanies the guidelines to aid in clinical practice and inform future audits of outcomes.
What are the main differences between MPRAs and STARR-seq?
5 answers
Massively parallel reporter assays (MPRAs) and STARR-seq are both high-throughput methods for assessing gene regulatory elements. MPRAs screen thousands of sequences simultaneously for regulatory activity, while STARR-seq quantitatively evaluates enhancers on a genome-wide scale using transcribed RNA. MPRAs have various experimental designs, including episomal and lentiviral approaches, influencing results subtly based on epigenetic and sequence features. In contrast, STARR-seq focuses on enhancer activity, with a recent study identifying over 33,000 candidate enhancers in HepG2 cells. While MPRAs are versatile for different questions in gene regulation, STARR-seq's strength lies in unbiased and quantitative assessment of enhancers at a genome-wide scale, emphasizing the importance of a reliable processing pipeline.
What are the specific molecular pathways involved in the activation and regulation of IL-17 signaling during inflammation?
10 answers
The activation and regulation of IL-17 signaling during inflammation involve a complex interplay of molecular pathways that orchestrate the immune response. IL-17 signaling is initiated when IL-17 cytokines bind to their receptor, IL-17R, leading to the recruitment of the adaptor molecule Act1. This interaction is crucial for the downstream activation of various signaling cascades, including the NF-κB and MAPK pathways, which are central to the inflammatory response. Act1 functions as a U-box E3 ubiquitin ligase, essential for IL-17 signaling, and its activity is regulated by phosphorylation through the kinase IKKi (IKKe), which is identified as a critical regulator of Act1 and IL-17 signaling. IKKi phosphorylates Act1, enhancing its ability to activate the MAPK pathway and stabilize chemokine mRNAs, thereby amplifying the inflammatory response. The IL-17 signaling pathway also involves the linear ubiquitin chain assembly complex (LUBAC) as an important signal transducing component of the IL-17R complex. IL-17 signaling is further regulated by a robust negative feedback loop mediated by the kinases TBK1 and IKKe, which terminate IL-17 signaling by phosphorylating the adaptor ACT1, leading to the release of the essential ubiquitin ligase TRAF6 from the complex. This regulatory mechanism is crucial for preventing excessive inflammation. Moreover, IL-17 interacts with other signaling molecules and pathways to exert its effects. For instance, IL-17 can enhance the expression of RANKL and inhibit OPG in human periodontal ligament cells, implicating it in bone remodeling processes during inflammation. Additionally, IL-17 can modulate the expression of genes involved in the JAK/STAT pathway, influencing the differentiation of Th17 cells and the inflammatory response. Furthermore, IL-17-induced gene expression is modulated by posttranscriptional mechanisms, including the stabilization of mRNA by the RNA binding protein Arid5a, which integrates IL-17-driven signaling pathways through posttranscriptional control. This highlights the multifaceted nature of IL-17 signaling regulation, involving both transcriptional and posttranscriptional mechanisms to fine-tune the inflammatory response. In summary, the activation and regulation of IL-17 signaling during inflammation involve a network of molecular pathways, including the NF-κB, MAPK, and JAK/STAT pathways, regulated by key molecules such as Act1, IKKi, TBK1, and Arid5a, which together ensure a balanced immune response.
How is a full blood count undertaken in a laboratory?
5 answers
A full blood count (FBC) in a laboratory involves measuring various parameters like red blood cell count, white blood cell count, platelet count, hemoglobin levels, and red cell parameters. Different instruments, such as aperture impedance or light scattering-based analyzers, are used for blood counting. These instruments can be semi-automated or fully automated multi-channel analyzers, with the latter offering more advanced features like internal dilution systems and automatic specimen handling. The process includes dilution, measurement of red cell parameters, and calibration to ensure accurate results and harmonization across different instruments. Additionally, microfluidic devices have been developed for FBC testing, incorporating channels for different measurements and solutions like lysis agents and quench solutions. Understanding FBC procedures is crucial for healthcare assistants and professionals to provide effective patient care.