Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Blood ; 140(19): 2037-2052, 2022 11 10.
Article in English | MEDLINE | ID: mdl-35984907

ABSTRACT

Targeting altered tumor cell metabolism might provide an attractive opportunity for patients with acute myeloid leukemia (AML). An amino acid dropout screen on primary leukemic stem cells and progenitor populations revealed a number of amino acid dependencies, of which methionine was one of the strongest. By using various metabolite rescue experiments, nuclear magnetic resonance-based metabolite quantifications and 13C-tracing, polysomal profiling, and chromatin immunoprecipitation sequencing, we identified that methionine is used predominantly for protein translation and to provide methyl groups to histones via S-adenosylmethionine for epigenetic marking. H3K36me3 was consistently the most heavily impacted mark following loss of methionine. Methionine depletion also reduced total RNA levels, enhanced apoptosis, and induced a cell cycle block. Reactive oxygen species levels were not increased following methionine depletion, and replacement of methionine with glutathione or N-acetylcysteine could not rescue phenotypes, excluding a role for methionine in controlling redox balance control in AML. Although considered to be an essential amino acid, methionine can be recycled from homocysteine. We uncovered that this is primarily performed by the enzyme methionine synthase and only when methionine availability becomes limiting. In vivo, dietary methionine starvation was not only tolerated by mice, but also significantly delayed both cell line and patient-derived AML progression. Finally, we show that inhibition of the H3K36-specific methyltransferase SETD2 phenocopies much of the cytotoxic effects of methionine depletion, providing a more targeted therapeutic approach. In conclusion, we show that methionine depletion is a vulnerability in AML that can be exploited therapeutically, and we provide mechanistic insight into how cells metabolize and recycle methionine.


Subject(s)
Leukemia, Myeloid, Acute , Methionine , Mice , Animals , Leukemia, Myeloid, Acute/pathology , S-Adenosylmethionine/metabolism , S-Adenosylmethionine/therapeutic use , Histones/metabolism , Racemethionine
2.
Blood Adv ; 6(7): 2129-2143, 2022 04 12.
Article in English | MEDLINE | ID: mdl-34543390

ABSTRACT

Acute myeloid leukemia (AML) often presents as an oligoclonal disease whereby multiple genetically distinct subclones can coexist within patients. Differences in signaling and drug sensitivity of such subclones complicate treatment and warrant tools to identify them and track disease progression. We previously identified >50 AML-specific plasma membrane (PM) proteins, and 7 of these (CD82, CD97, FLT3, IL1RAP, TIM3, CD25, and CD123) were implemented in routine diagnostics in patients with AML (n = 256) and myelodysplastic syndrome (n = 33). We developed a pipeline termed CombiFlow in which expression data of multiple PM markers is merged, allowing a principal component-based analysis to identify distinctive marker expression profiles and to generate single-cell t-distributed stochastic neighbor embedding landscapes to longitudinally track clonal evolution. Positivity for one or more of the markers after 2 courses of intensive chemotherapy predicted a shorter relapse-free survival, supporting a role for these markers in measurable residual disease (MRD) detection. CombiFlow also allowed the tracking of clonal evolution in paired diagnosis and relapse samples. Extending the panel to 36 AML-specific markers further refined the CombiFlow pipeline. In conclusion, CombiFlow provides a valuable tool in the diagnosis, MRD detection, clonal tracking, and understanding of clonal heterogeneity in AML.


Subject(s)
Leukemia, Myeloid, Acute , Cell Membrane/metabolism , Clonal Evolution/genetics , Clone Cells/metabolism , Humans , Interleukin-3 Receptor alpha Subunit , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics
3.
Cell Stem Cell ; 26(5): 755-765.e7, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32386556

ABSTRACT

Hematopoietic stem cells (HSCs) require highly regulated rates of protein synthesis, but it is unclear if they or lineage-committed progenitors preferentially recruit transcripts to translating ribosomes. We utilized polysome profiling, RNA sequencing, and whole-proteomic approaches to examine the translatome in LSK (Lin-Sca-1+c-Kit+) and myeloid progenitor (MP; Lin-Sca-1-c-Kit+) cells. Our studies show that LSKs exhibit low global translation but high translational efficiencies (TEs) of mRNAs required for HSC maintenance. In contrast, MPs activate translation in an mTOR-independent manner due, at least in part, to proteasomal degradation of mTOR by the E3 ubiquitin ligase c-Cbl. In the near absence of mTOR, CDK1 activates eIF4E-dependent translation in MPs through phosphorylation of 4E-BP1. Aberrant activation of mTOR expression and signaling in c-Cbl-deficient MPs results in increased mature myeloid lineage output. Overall, our data demonstrate that hematopoietic stem and progenitor cells (HSPCs) undergo translational reprogramming mediated by previously uncharacterized mechanisms of translational regulation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Proteomics , Hematopoietic Stem Cells , Signal Transduction , TOR Serine-Threonine Kinases
4.
J Exp Med ; 216(10): 2362-2377, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31371381

ABSTRACT

Despite significant efforts to improve therapies for acute myeloid leukemia (AML), clinical outcomes remain poor. Understanding the mechanisms that regulate the development and maintenance of leukemic stem cells (LSCs) is important to reveal new therapeutic opportunities. We have identified CD97, a member of the adhesion class of G protein-coupled receptors (GPCRs), as a frequently up-regulated antigen on AML blasts that is a critical regulator of blast function. High levels of CD97 correlate with poor prognosis, and silencing of CD97 reduces disease aggressiveness in vivo. These phenotypes are due to CD97's ability to promote proliferation, survival, and the maintenance of the undifferentiated state in leukemic blasts. Collectively, our data credential CD97 as a promising therapeutic target on LSCs in AML.


Subject(s)
Antigens, CD/biosynthesis , Blast Crisis/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Up-Regulation , Animals , Antigens, CD/genetics , Blast Crisis/genetics , Blast Crisis/pathology , Cell Proliferation , Cell Survival , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/genetics
5.
Cancer Cell ; 34(4): 674-689.e8, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30245083

ABSTRACT

Intra-tumor heterogeneity caused by clonal evolution is a major problem in cancer treatment. To address this problem, we performed label-free quantitative proteomics on primary acute myeloid leukemia (AML) samples. We identified 50 leukemia-enriched plasma membrane proteins enabling the prospective isolation of genetically distinct subclones from individual AML patients. Subclones differed in their regulatory phenotype, drug sensitivity, growth, and engraftment behavior, as determined by RNA sequencing, DNase I hypersensitive site mapping, transcription factor occupancy analysis, in vitro culture, and xenograft transplantation. Finally, we show that these markers can be used to identify and longitudinally track distinct leukemic clones in patients in routine diagnostics. Our study describes a strategy for a major improvement in stratifying cancer diagnosis and treatment.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Phenotype , Transcription Factors/genetics , Adult , Aged , Base Sequence/genetics , Clonal Evolution/genetics , Humans , Male , Middle Aged , Prospective Studies , fms-Like Tyrosine Kinase 3/genetics
6.
J Exp Med ; 214(8): 2453-2470, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28733386

ABSTRACT

The microRNA-99 (miR-99) family comprises a group of broadly conserved microRNAs that are highly expressed in hematopoietic stem cells (HSCs) and acute myeloid leukemia stem cells (LSCs) compared with their differentiated progeny. Herein, we show that miR-99 regulates self-renewal in both HSCs and LSCs. miR-99 maintains HSC long-term reconstitution activity by inhibiting differentiation and cell cycle entry. Moreover, miR-99 inhibition induced LSC differentiation and depletion in an MLL-AF9-driven mouse model of AML, leading to reduction in leukemia-initiating activity and improved survival in secondary transplants. Confirming miR-99's role in established AML, miR-99 inhibition induced primary AML patient blasts to undergo differentiation. A forward genetic shRNA library screen revealed Hoxa1 as a critical mediator of miR-99 function in HSC maintenance, and this observation was independently confirmed in both HSCs and LSCs. Together, these studies demonstrate the importance of noncoding RNAs in the regulation of HSC and LSC function and identify miR-99 as a critical regulator of stem cell self-renewal.


Subject(s)
Cell Self Renewal/genetics , Cell Self Renewal/physiology , Hematopoietic Stem Cells/physiology , MicroRNAs/genetics , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Transcription Factors/genetics
7.
Blood ; 129(1): 48-59, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27827827

ABSTRACT

K(lysine) acetyltransferase 8 (KAT8, also known as MOF) mediates the acetylation of histone H4 at lysine 16 (H4K16ac) and is crucial for murine embryogenesis. Lysine acetyltransferases have been shown to regulate various stages of normal hematopoiesis. However, the function of MOF in hematopoietic stem cell (HSC) development has not yet been elucidated. We set out to study the role of MOF in general hematopoiesis by using a Vav1-cre-induced conditional murine Mof knockout system and found that MOF is critical for hematopoietic cell maintenance and HSC engraftment capacity in adult hematopoiesis. Rescue experiments with a MOF histone acetyltransferase domain mutant illustrated the requirement for MOF acetyltransferase activity in the clonogenic capacity of HSCs and progenitors. In stark contrast, fetal steady-state hematopoiesis at embryonic day (E) 14.5 was not affected by homozygous Mof deletion despite dramatic loss of global H4K16ac. Hematopoietic defects start manifesting in late gestation at E17.5. The discovery that MOF and its H4K16ac activity are required for adult but not early and midgestational hematopoiesis supports the notion that multiple chromatin regulators may be crucial for hematopoiesis at varying stages of development. MOF is therefore a developmental-stage-specific chromatin regulator found to be essential for adult but not early fetal hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Histone Acetyltransferases/metabolism , Animals , Fetus , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
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
9.
Blood ; 127(10): 1242-8, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26787736

ABSTRACT

The classical model of hematopoiesis has long held that hematopoietic stem cells (HSCs) sit at the apex of a developmental hierarchy in which HSCs undergo long-term self-renewal while giving rise to cells of all the blood lineages. In this model, self-renewing HSCs progressively lose the capacity for self-renewal as they transit into short-term self-renewing and multipotent progenitor states, with the first major lineage commitment occurring in multipotent progenitors, thus giving rise to progenitors that initiate the myeloid and lymphoid branches of hematopoiesis. Subsequently, within the myeloid lineage, bipotent megakaryocyte-erythrocyte and granulocyte-macrophage progenitors give rise to unipotent progenitors that ultimately give rise to all mature progeny. However, over the past several years, this developmental scheme has been challenged, with the origin of megakaryocyte precursors being one of the most debated subjects. Recent studies have suggested that megakaryocytes can be generated from multiple pathways and that some differentiation pathways do not require transit through a requisite multipotent or bipotent megakaryocyte-erythrocyte progenitor stage. Indeed, some investigators have argued that HSCs contain a subset of cells with biased megakaryocyte potential, with megakaryocytes directly arising from HSCs under steady-state and stress conditions. In this review, we discuss the evidence supporting these nonclassical megakaryocytic differentiation pathways and consider their relative strengths and weaknesses as well as the technical limitations and potential pitfalls in interpreting these studies. Ultimately, such pitfalls will need to be overcome to provide a comprehensive and definitive understanding of megakaryopoiesis.


Subject(s)
Cell Differentiation/physiology , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/metabolism , Myelopoiesis/physiology , Thrombopoiesis/physiology , Animals , Humans , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis/physiology , Megakaryocyte-Erythroid Progenitor Cells/cytology , Megakaryocytes/cytology
10.
Biol Blood Marrow Transplant ; 20(6): 865-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24607555

ABSTRACT

Most of our knowledge of the effects of aging on the hematopoietic system comes from studies in animal models. In this study, to explore potential effects of aging on human hematopoietic stem and progenitor cells (HSPCs), we evaluated CD34(+) cells derived from young (<35 years) and old (>60 years) adult bone marrow with respect to phenotype and in vitro function. We observed an increased frequency of phenotypically defined stem and progenitor cells with age, but no distinct differences with respect to in vitro functional capacity. Given that regeneration of peripheral blood counts can serve as a functional readout of HSPCs, we compared various peripheral blood parameters between younger patients (≤50 years; n = 64) and older patients (≥60 years; n = 55) after autologous stem cell transplantation. Patient age did not affect the number of apheresis cycles or the amount of CD34(+) cells harvested. Parameters for short-term regeneration did not differ significantly between the younger and older patients; however, complete recovery of all 3 blood lineages at 1 year after transplantation was strongly affected by advanced age, occurring in only 29% of the older patients, compared with 56% of the younger patients (P = .009). Collectively, these data suggest that aging has only limited effects on CD34(+) HSPCs under steady-state conditions, but can be important under consitions of chemotoxic and replicative stress.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Adult , Age Factors , Antigens, CD34/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cohort Studies , Female , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/therapy , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/therapy , Transplantation, Autologous , Young Adult
11.
Haematologica ; 98(12): 1964-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038022

ABSTRACT

Patients who have undergone autologous stem cell transplantation are subsequently more susceptible to chemotherapy-induced bone marrow toxicity. In the present study, bone marrow primitive progenitor cells were examined one year after autologous stem cell transplantation and compared with normal bone marrow and mobilized peripheral blood stem cells. Post-transplantation bone marrow contained a significantly lower percentage of quiescent cells in the CD34(+)/CD38(low) fraction compared to normal bone marrow. In addition, we observed a strong decrease in stem cell/primitive progenitor frequency in post-transplantation CD34(+) cells as defined by long-term culture assays. Measurement of the levels of reactive oxygen species by flow cytometry revealed comparable levels in post-transplantation and normal bone marrow CD34(+)/CD38(low) cells, while significantly higher levels of reactive oxygen species were observed in CD34(+)/CD38(high) cells following autologous stem cell transplantation compared to normal bone marrow. Moreover, post-transplantation CD34(+) bone marrow cells demonstrated an increased sensitivity to buthionine sulfoximine, a trigger for endogenous production of reactive oxygen species. Gene expression analysis on CD34(+) cells revealed a set of 195 genes, including HMOX1, EGR1, FOS and SIRPA that are persistently down-regulated in mobilized peripheral blood cells and post-transplantation bone marrow compared to normal bone marrow. In conclusion, our data indicate that the diminished regenerative capacity of bone marrow following autologous stem cell transplantation is possibly related to a loss of quiescence and a reduced tolerability to oxidative stress.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Antigens, CD34/physiology , Hematopoietic Stem Cell Transplantation/trends , Adult , Aged , Cells, Cultured , Female , Follow-Up Studies , Humans , Male , Middle Aged , Reactive Oxygen Species/metabolism , Time Factors , Transplantation, Autologous/trends
12.
Haematologica ; 98(10): 1532-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23716555

ABSTRACT

Mutations of nucleophosmin 1 are frequently found in acute myeloid leukemia and lead to aberrant cytoplasmic accumulation of nucleophosmin protein. Immunohistochemical staining is therefore recommended as the technique of choice in front-line screening. In this study, we assessed the sensitivity and specificity of immunohistochemistry on formalin-fixed bone marrow biopsies compared with gold standard molecular analysis to predict nucleophosmin 1 mutation status in 119 patients with acute myeloid leukemia. Discrepant cases were further characterized by gene expression analyses and fluorescence in situ hybridization. A large overlap between both methods was observed. Nevertheless, nine patients demonstrated discordant results at initial screening. Five cases demonstrated nuclear staining of nucleophosmin 1 by immunohistochemistry, but a nucleophosmin 1 mutation by molecular analysis. In two cases this could be attributed to technical issues and in three cases minor subpopulations of myeloblasts had not been discovered initially. All tested cases exhibited the characteristic nucleophosmin-mutated gene expression pattern. Four cases had cytoplasmic nucleophosmin 1 staining and a nucleophosmin-mutated gene expression pattern without a detectable nucleophosmin 1 mutation. In two of these cases we found the chromosomal translocation t(3;5)(q25;q35) encoding the NPM-MLF1 fusion protein. In the other discrepant cases the aberrant cytoplasmic nucleophosmin staining and gene expression could not be explained. In total six patients (5%) had true discordant results between immunohistochemistry and mutation analysis. We conclude that cytoplasmic nucleophosmin localization is not always caused by a conventional nucleophosmin 1 mutation and that in the screening for nucleophosmin 1 abnormalities, most information will be obtained by combining immunohistochemistry with molecular analysis.


Subject(s)
DNA Mutational Analysis/methods , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Nuclear Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nucleophosmin , Young Adult
13.
Curr Opin Immunol ; 23(4): 512-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21664115

ABSTRACT

During development hematopoietic stem cells (HSCs) expand in number and persist throughout life by undergoing self-renewing divisions. Nevertheless, the hematopoietic system does not escape the negative effects of aging, suggesting that self-renewal is not complete. A fundamental issue in stem cell biology relates to such age-dependent loss of stem cell activity. Both stem cell intrinsic factors and extrinsic factors associated with an aging micro-environment could contribute to aging of the hematopoietic system. Recently, changes in the clonal composition of the HSC compartment during aging have been put forward as a key factor. Here, we discuss these recent developments and speculate how they may be of clinical relevance.


Subject(s)
Aging/immunology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic System/growth & development , Stem Cell Niche/physiology , Age of Onset , Aged , Aging/blood , Animals , Bone Marrow/growth & development , Cell Lineage , Clone Cells/cytology , Gene Expression Regulation, Developmental , Hematopoietic System/cytology , Humans , Leukemia, Myeloid/epidemiology , Leukemia, Myeloid/pathology , Lymphocytes/cytology , Mice , Models, Immunological , Myeloid Cells/cytology , Stromal Cells/physiology
14.
Blood ; 114(14): 2869-77, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19667402

ABSTRACT

Acute myeloid leukemia (AML) has a different clinical and biologic behavior in patients at older age. To gain further insight into the molecular differences, we examined a cohort of 525 adults to compare gene expression profiles of the one-third of youngest cases (n = 175; median age 31 years) with the one-third of oldest cases (n = 175; median age 59 years). This analysis revealed that 477 probe sets were up-regulated and 492 probe sets were down-regulated with increasing age at the significance level of P < .00001. After validation with 2 independent AML cohorts, the 969 differentially regulated probe sets on aging could be pointed to 41 probe sets, including the tumor-suppressor gene CDKN2A (encoding p16(INK4A)). In contrast to the induced p16(INK4A) expression that is associated with physiologic aging, p16(INK4A) is down-regulated in AML samples of patients with increasing age. However, this was only noticed in the intermediate- and unfavorable-risk group and not in the favorable-risk group and the molecularly defined subset "NPM1 mutant without FLT3-ITD." Multivariate analysis revealed p16(INK4A), besides cytogenetic risk groups, as an independent prognostic parameter for overall survival in older patients. We conclude that, in addition to altered clinical and biologic characteristics, AML presenting at older age shows different gene expression profiles.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , RNA, Messenger/genetics , Adolescent , Adult , Age Factors , Aged , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Middle Aged , Nucleophosmin , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Young Adult
15.
Aging (Albany NY) ; 1(11): 949-53, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-20157576

ABSTRACT

Aging is generally considered to be the consequence of stem cell attrition caused by the activity of tumor suppressor pathways that censor potentially malignant clones by eliciting apoptosis or senescence. An important effector of aging is the cyclindependent kinase inhibitor p16(INK4a), which is also a known suppressor of cancer. The expression of p16(INK4a) is very low or absent in young organisms but increases with advancing age. We recently showed that, unlike healthy cells, acute myeloid leukemia (AML) derived blasts show a down-regulation of p16(INK4a) mRNA with increasing age. Based on this observation we hypothesize that suppression of defense mechanisms which protect older cells against cellular and DNA damage might facilitate oncogenesis in older individuals.


Subject(s)
Aging/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Down-Regulation/genetics , Leukemia, Myeloid, Acute/genetics , RNA, Messenger/genetics , Aging/pathology , Aging/physiology , Apoptosis/genetics , Apoptosis/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , DNA Damage/genetics , DNA Damage/physiology , Down-Regulation/physiology , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/physiopathology , Leukocytes/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...