Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 10(4): 536-551, 2020 04.
Article in English | MEDLINE | ID: mdl-31974170

ABSTRACT

Venetoclax-based therapy can induce responses in approximately 70% of older previously untreated patients with acute myeloid leukemia (AML). However, up-front resistance as well as relapse following initial response demonstrates the need for a deeper understanding of resistance mechanisms. In the present study, we report that responses to venetoclax +azacitidine in patients with AML correlate closely with developmental stage, where phenotypically primitive AML is sensitive, but monocytic AML is more resistant. Mechanistically, resistant monocytic AML has a distinct transcriptomic profile, loses expression of venetoclax target BCL2, and relies on MCL1 to mediate oxidative phosphorylation and survival. This differential sensitivity drives a selective process in patients which favors the outgrowth of monocytic subpopulations at relapse. Based on these findings, we conclude that resistance to venetoclax + azacitidine can arise due to biological properties intrinsic to monocytic differentiation. We propose that optimal AML therapies should be designed so as to independently target AML subclones that may arise at differing stages of pathogenesis. SIGNIFICANCE: Identifying characteristics of patients who respond poorly to venetoclax-based therapy and devising alternative therapeutic strategies for such patients are important topics in AML. We show that venetoclax resistance can arise due to intrinsic molecular/metabolic properties of monocytic AML cells and that such properties can potentially be targeted with alternative strategies.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/therapeutic use , Aged , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Humans , Sulfonamides/pharmacology
2.
Blood ; 134(4): 389-394, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31101624

ABSTRACT

We have previously demonstrated that oxidative phosphorylation is required for the survival of human leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML). More recently, we demonstrated that LSCs in patients with de novo AML rely on amino acid metabolism to drive oxidative phosphorylation. Notably, although overall levels of amino acids contribute to LSC energy metabolism, our current findings suggest that cysteine may be of particular importance for LSC survival. We demonstrate that exogenous cysteine is metabolized exclusively to glutathione. Upon cysteine depletion, glutathione synthesis is impaired, leading to reduced glutathionylation of succinate dehydrogenase A (SDHA), a key component of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation impairs ETC II activity, thereby inhibiting oxidative phosphorylation, reducing production of ATP, and leading to LSC death. Given the role of cysteine in driving LSC energy production, we tested cysteine depletion as a potential therapeutic strategy. Using a novel cysteine-degrading enzyme, we demonstrate selective eradication of LSCs, with no detectable effect on normal hematopoietic stem/progenitor cells. Together, these findings indicate that LSCs are aberrantly reliant on cysteine to sustain energy metabolism, and that targeting this axis may represent a useful therapeutic strategy.


Subject(s)
Cysteine/metabolism , Electron Transport Complex II/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Biomarkers , Energy Metabolism , Glutathione/metabolism , Humans , Oxidation-Reduction , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism
3.
Cell Rep ; 27(1): 238-254.e6, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943405

ABSTRACT

The NADPH-dependent oxidase NOX2 is an important effector of immune cell function, and its activity has been linked to oncogenic signaling. Here, we describe a role for NOX2 in leukemia-initiating stem cell populations (LSCs). In a murine model of leukemia, suppression of NOX2 impaired core metabolism, attenuated disease development, and depleted functionally defined LSCs. Transcriptional analysis of purified LSCs revealed that deficiency of NOX2 collapses the self-renewal program and activates inflammatory and myeloid-differentiation-associated programs. Downstream of NOX2, we identified the forkhead transcription factor FOXC1 as a mediator of the phenotype. Notably, suppression of NOX2 or FOXC1 led to marked differentiation of leukemic blasts. In xenotransplantation models of primary human myeloid leukemia, suppression of either NOX2 or FOXC1 significantly attenuated disease development. Collectively, these findings position NOX2 as a critical regulator of malignant hematopoiesis and highlight the clinical potential of inhibiting NOX2 as a means to target LSCs.


Subject(s)
Cell Self Renewal , Leukemia/blood , Leukopoiesis , Myeloid Progenitor Cells/metabolism , NADPH Oxidase 2/metabolism , Animals , Cells, Cultured , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Leukemia/genetics , Leukemia/metabolism , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/pathology , NADPH Oxidase 2/genetics
5.
Cancer Cell ; 34(5): 724-740.e4, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423294

ABSTRACT

In this study we interrogated the metabolome of human acute myeloid leukemia (AML) stem cells to elucidate properties relevant to therapeutic intervention. We demonstrate that amino acid uptake, steady-state levels, and catabolism are all elevated in the leukemia stem cell (LSC) population. Furthermore, LSCs isolated from de novo AML patients are uniquely reliant on amino acid metabolism for oxidative phosphorylation and survival. Pharmacological inhibition of amino acid metabolism reduces oxidative phosphorylation and induces cell death. In contrast, LSCs obtained from relapsed AML patients are not reliant on amino acid metabolism due to their ability to compensate through increased fatty acid metabolism. These findings indicate that clinically relevant eradication of LSCs can be achieved with drugs that target LSC metabolic vulnerabilities.


Subject(s)
Amino Acids/metabolism , Fatty Acids/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Oxidative Phosphorylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Biological Transport/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Female , Glycolysis/physiology , Humans , Leukemia, Myeloid, Acute/drug therapy , Lipid Metabolism/drug effects , Metabolome/physiology , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology
6.
Cancer Cell ; 34(4): 659-673.e6, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30270124

ABSTRACT

From an organismal perspective, cancer cell populations can be considered analogous to parasites that compete with the host for essential systemic resources such as glucose. Here, we employed leukemia models and human leukemia samples to document a form of adaptive homeostasis, where malignant cells alter systemic physiology through impairment of both host insulin sensitivity and insulin secretion to provide tumors with increased glucose. Mechanistically, tumor cells induce high-level production of IGFBP1 from adipose tissue to mediate insulin sensitivity. Further, leukemia-induced gut dysbiosis, serotonin loss, and incretin inactivation combine to suppress insulin secretion. Importantly, attenuated disease progression and prolonged survival are achieved through disruption of the leukemia-induced adaptive homeostasis. Our studies provide a paradigm for systemic management of leukemic disease.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Insulin Resistance/physiology , Leukemia/metabolism , Animals , Diet, High-Fat , Humans , Insulin/biosynthesis , Mice
7.
Nat Commun ; 9(1): 3694, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209285

ABSTRACT

Myelodysplastic syndrome (MDS) is a chronic hematologic disorder that frequently evolves to more aggressive stages and in some cases leads to acute myeloid leukemia (AML). MDS arises from mutations in hematopoietic stem cells (HSCs). Thus, to define optimal therapies, it is essential to understand molecular events driving HSC pathogenesis. In this study, we report that during evolution of MDS, malignant HSCs activate distinct cellular programs that render such cells susceptible to therapeutic intervention. Specifically, metabolic analyses of the MDS stem cell compartment show a profound activation of protein synthesis machinery and increased oxidative phosphorylation. Pharmacological targeting of protein synthesis and oxidative phosphorylation demonstrated potent and selective eradication of MDS stem cells in primary human patient specimens. Taken together, our findings indicate that MDS stem cells are reliant on specific metabolic events and that such properties can be targeted prior to the onset of clinically significant AML, during antecedent MDS.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Hematopoietic Stem Cells/metabolism , Myelodysplastic Syndromes/metabolism , Cells, Cultured , Flow Cytometry , Humans , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/metabolism , Oxidative Phosphorylation
8.
Cell Stem Cell ; 23(1): 86-100.e6, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29910151

ABSTRACT

Leukemia stem cells (LSCs) are thought to drive the genesis of acute myeloid leukemia (AML) as well as relapse following chemotherapy. Because of their unique biology, developing effective methods to eradicate LSCs has been a significant challenge. In the present study, we demonstrate that intrinsic overexpression of the mitochondrial dynamics regulator FIS1 mediates mitophagy activity that is essential for primitive AML cells. Depletion of FIS1 attenuates mitophagy and leads to inactivation of GSK3, myeloid differentiation, cell cycle arrest, and a profound loss of LSC self-renewal potential. Further, we report that the central metabolic stress regulator AMPK is also intrinsically activated in LSC populations and is upstream of FIS1. Inhibition of AMPK signaling recapitulates the biological effect of FIS1 loss. These data suggest a model in which LSCs co-opt AMPK/FIS1-mediated mitophagy as a means to maintain stem cell properties that may be otherwise compromised by the stresses induced by oncogenic transformation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Self Renewal , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Neoplastic Stem Cells/pathology , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Mitophagy/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
9.
Haematologica ; 102(6): 1054-1065, 2017 06.
Article in English | MEDLINE | ID: mdl-28280079

ABSTRACT

Aldehyde dehydrogenase 1A1 (ALDH1A1) activity is high in hematopoietic stem cells and functions in part to protect stem cells from reactive aldehydes and other toxic compounds. In contrast, we found that approximately 25% of all acute myeloid leukemias expressed low or undetectable levels of ALDH1A1 and that this ALDH1A1- subset of leukemias correlates with good prognosis cytogenetics. ALDH1A1- cell lines as well as primary leukemia cells were found to be sensitive to treatment with compounds that directly and indirectly generate toxic ALDH substrates including 4-hydroxynonenal and the clinically relevant compounds arsenic trioxide and 4-hydroperoxycyclophosphamide. In contrast, normal hematopoietic stem cells were relatively resistant to these compounds. Using a murine xenotransplant model to emulate a clinical treatment strategy, established ALDH1A1- leukemias were also sensitive to in vivo treatment with cyclophosphamide combined with arsenic trioxide. These results demonstrate that targeting ALDH1A1- leukemic cells with toxic ALDH1A1 substrates such as arsenic and cyclophosphamide may be a novel targeted therapeutic strategy for this subset of acute myeloid leukemias.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Drug Therapy, Combination/methods , Leukemia, Myeloid, Acute/drug therapy , Aldehyde Dehydrogenase 1 Family , Animals , Arsenic Trioxide , Arsenicals/therapeutic use , Cells, Cultured , Cyclophosphamide/therapeutic use , Heterografts , Humans , Leukemia, Myeloid, Acute/enzymology , Mice , Molecular Targeted Therapy , Oxides/therapeutic use , Retinal Dehydrogenase
11.
J Biol Chem ; 291(42): 21984-22000, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27573247

ABSTRACT

Although multidrug approaches to cancer therapy are common, few strategies are based on rigorous scientific principles. Rather, drug combinations are largely dictated by empirical or clinical parameters. In the present study we developed a strategy for rational design of a regimen that selectively targets human acute myelogenous leukemia (AML) stem cells. As a starting point, we used parthenolide, an agent shown to target critical mechanisms of redox balance in primary AML cells. Next, using proteomic, genomic, and metabolomic methods, we determined that treatment with parthenolide leads to induction of compensatory mechanisms that include up-regulated NADPH production via the pentose phosphate pathway as well as activation of the Nrf2-mediated oxidative stress response pathway. Using this knowledge we identified 2-deoxyglucose and temsirolimus as agents that can be added to a parthenolide regimen as a means to inhibit such compensatory events and thereby further enhance eradication of AML cells. We demonstrate that the parthenolide, 2-deoxyglucose, temsirolimus (termed PDT) regimen is a potent means of targeting AML stem cells but has little to no effect on normal stem cells. Taken together our findings illustrate a comprehensive approach to designing combination anticancer drug regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Deoxyglucose/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , NADP/biosynthesis , Neoplastic Stem Cells/pathology , Sesquiterpenes/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Up-Regulation/drug effects
12.
Cell Stem Cell ; 19(1): 23-37, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27374788

ABSTRACT

Adipose tissue (AT) has previously been identified as an extra-medullary reservoir for normal hematopoietic stem cells (HSCs) and may promote tumor development. Here, we show that a subpopulation of leukemic stem cells (LSCs) can utilize gonadal adipose tissue (GAT) as a niche to support their metabolism and evade chemotherapy. In a mouse model of blast crisis chronic myeloid leukemia (CML), adipose-resident LSCs exhibit a pro-inflammatory phenotype and induce lipolysis in GAT. GAT lipolysis fuels fatty acid oxidation in LSCs, especially within a subpopulation expressing the fatty acid transporter CD36. CD36(+) LSCs have unique metabolic properties, are strikingly enriched in AT, and are protected from chemotherapy by the GAT microenvironment. CD36 also marks a fraction of human blast crisis CML and acute myeloid leukemia (AML) cells with similar biological properties. These findings suggest striking interplay between leukemic cells and AT to create a unique microenvironment that supports the metabolic demands and survival of a distinct LSC subpopulation.


Subject(s)
Adaptation, Physiological , Adipose Tissue/pathology , Antineoplastic Agents/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/therapeutic use , Blast Crisis/drug therapy , Blast Crisis/pathology , CD36 Antigens/metabolism , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Energy Metabolism/drug effects , Fatty Acids/metabolism , Gonads/pathology , Humans , Inflammation/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Lipolysis/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/drug effects , Oxidation-Reduction/drug effects , Tumor Burden/drug effects
13.
J Health Popul Nutr ; 30(1): 22-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22524116

ABSTRACT

Internalized stigma among people living with HIV/AIDS (PLHA) is prevalent in Bangladesh. A better understanding of the effects of stigma on PLHA is required to reduce this and to minimize its harmful effects. This study employed a quantitative approach by conducting a survey with an aim to know the prevalence of internalized stigma and to identify the factors associated with internalized stigma among a sample of 238 PLHA (male=152 and female=86) in Bangladesh. The findings suggest that there is a significant difference between groups with the low- and the high-internalized HIV/AIDS stigma in terms of both age and gender. The prevalence of internalized stigma varied according to the poverty status of PLHA. An exploratory factor analysis (EFA) found 10 of 15 items loaded highly on the three factors labelled self-acceptance, self-exclusion, and social withdrawal. About 68% of the PLHA felt ashamed, and 54% felt guilty because of their HIV status. More than half (87.5% male and 19.8% female) of the PLHA blamed themselves for their HIV status while many of them (38.2% male and 8.1% female) felt that they should be punished. The male PLHA more frequently chose to withdraw themselves from family and social gatherings compared to the female PLHA. They also experienced a higher level of internalized stigma compared to the female PLHA. The results suggest that the prevalence of internalized stigma is high in Bangladesh, and much needs to be done by different organizations working for and with the PLHA to reduce internalized stigma among this vulnerable group.


Subject(s)
Guilt , HIV Infections/psychology , Self Concept , Stereotyping , Acquired Immunodeficiency Syndrome/psychology , Adolescent , Adult , Attitude to Health , Bangladesh , Factor Analysis, Statistical , Female , Humans , Male , Middle Aged , Poverty , Sex Distribution , Social Stigma , Surveys and Questionnaires , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...