Rare Hematopoietic Subclones Harboring Leukemogenic TP53 Mutations Are Detectable Via Error-Corrected Sequencing in Healthy Elderly Individuals

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2907-2907
Author(s):  
Andrew Young ◽  
Terrence Neal Wong ◽  
Timothy J Ley ◽  
Daniel C. Link ◽  
Todd E Druley

Abstract Acute myeloid leukemia (AML) is an oligoclonal disease marked by specific somatic genomic alterations. While the leukemia-associated mutations and rearrangements differ between individual cases, the set of recurrently mutated genes is now largely known (Cancer Genome Atlas Research Network, NEJM 2013). Current evidence supports a model of leukemogenesis, by which leukemia-associated mutations are acquired sequentially over time in hematopoietic stem cells (HSCs). Furthermore, “pre-leukemic” HSCs, which contain only a subset of the mutations found in the dominant clone, are detectable at diagnosis (Corces-Zimmerman MR, et al., PNAS 2014; Shlush LI, et al., Nature 2014). Despite these observations, the effect of these mutations, when they first arise in healthy HSCs, is largely unknown. It is likely that these early mutations endow a selective growth advantage to the HSC resulting in detectable clonal hematopoiesis without immediately causing overt leukemia. As expected, there is evidence from studies of X-inactivation skewing that clonal hematopoiesis exists in the blood of healthy elderly individuals (Busque L, et al. Blood 2009). In a separate study, hematopoietic X-inactivation skewing in elderly individuals was associated with TET2 mutations in 10/182 cases (Busque L, et al. Nat Genet 2012). This study was only capable of detecting insertions or deletions due to the high (~1%) substitution error rate of conventional next-generation sequencing (NGS) and likely underreported the prevalence of clonal hematopoiesis harboring putative driver mutations in TET2. To further study the role of leukemia-associated single nucleotide variants in healthy hematopoiesis, we applied our validated method for targeted error-corrected sequencing (ECS). ECS uses random, single molecule indexing to overcome the inherent error rate of NGS by establishing “read families” from multiple reads generated from each unique index (Schmitt MW, et al. PNAS 2012, Kinde I, et al., PNAS 2012). A dilution series of two independent mutations with technical replicates demonstrated that ECS enables the quantitative identification of variants as rare as 1:10,000 molecules. We applied ECS to identify and quantify leukemia-associated subclones harboring mutations in TP53 exons 4-7, which is where the majority of cancer-related mutations in TP53 have been described. ECS libraries were generated from blood samples drawn from 20 healthy elderly individuals (average 75 years old). Sample multiplexing for sequencing was accomplished by tagging PCR amplicons, generated from each individual, with a different oligonucleotide barcode during library preparation. The resulting individual ECS libraries were then multiplexed and sequenced on one lane of the Illumina HiSeq 2500 platform. Sequence reads originating from the same randomly indexed molecule are aligned to each other to generate read families. First, at every position, the bases called by each sequence read are compared and a consensus base is called if there is ≥90% agreement between the reads. If there is less than 90% agreement, the consensus base is called an N. Sequencing errors are thus removed since they are not shared between different reads within a read family. Second, an error corrected consensus sequence (ECCS) is discarded if <90% of bases across a paired-end read are non-N. ECCSs are locally aligned to hg19/GRCh30 using bowtie2. We identified rare subclonal hematopoiesis harboring TP53 mutations in 9 of 20 healthy individuals at variant allele frequencies (VAF) between 1:10,000 and 1:270. Of the 13 identified mutations, 12 were coding or splicing mutations and 10 had been previously identified as leukemia-associated in the Catalog of Somatic Mutations in Cancer. We validated three independent variants with droplet digital PCR and measured nearly identical VAFs at each loci. These findings suggest that potentially oncogenic mutation in hematopoietic stem cells is a stochastic process and rare subclonal hematopoiesis is a common occurrence in healthy aged individuals, which is consistent with the observation that de novo AML primarily occurs in the elderly. Ongoing studies are applying ECS to determine the prevalence of rare subclonal mutation in other recurrently mutated AML genes. These studies will help further elucidate the natural history of leukemogenesis and may enable the accurate detection of individuals at risk for developing cancer. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (8) ◽  
pp. 3186-3193 ◽  
Author(s):  
Sabina I. Swierczek ◽  
Neeraj Agarwal ◽  
Roberto H. Nussenzveig ◽  
Gerald Rothstein ◽  
Andrew Wilson ◽  
...  

Abstract Clonality assays, based on X-chromosome inactivation, discriminate active from inactive alleles. Skewing of X-chromosome allelic usage, based on preferential methylation of one of the HUMARA alleles, was reported as evidence of clonal hematopoiesis in approximately 30% of elderly women. Using a quantitative, transcriptionally based clonality assay, we reported X-chromosome–transcribed allelic ratio in blood cells of healthy women consistent with random X-inactivation of 8 embryonic hematopoietic stem cells. Furthermore, we did not detect clonal hematopoiesis in more than 200 healthy nonelderly women. In view of the susceptibility of aging hematopoietic stem cells to epigenetic dysregulation, we reinvestigated the issue of clonality in elderly women. Forty healthy women (ages 65-92 years; mean, 81.3 years) were tested by a novel, quantitative polymerase chain reaction (qPCR) transcriptional clonality assay. We did not detect clonal hematopoiesis in any of the tested subjects. We also tested DNA from the same granulocyte samples using the methylation-based HUMARA assay, and confirmed previous reports of approximately 30% extensively skewed or monoallelic methylation, in agreement with likely age-related deregulated methylation of the HUMARA gene locus. We conclude that the transcriptionally based X-chromosome clonality assays are suitable for evaluation of clonal hematopoiesis in elderly women.


Circulation ◽  
2020 ◽  
Vol 142 (Suppl_3) ◽  
Author(s):  
Yoshimitsu Yura ◽  
Emiri Miura-Yura ◽  
Kenneth Walsh

Background: Therapy-related clonal hematopoiesis in cancer patients is typically associated with somatic mutations in hematopoietic cell genes that encode regulators of the DNA-damage response (DDR) pathway. The Protein Phosphatase Mg2+/Mn2+ Dependent 1D ( PPM1D ) gene is the most frequently mutated DDR gene associated with therapy-related clonal hematopoiesis. While epidemiological evidence suggests an association between therapy-related clonal hematopoiesis and cardiovascular disease in cancer patients, causal and mechanistic relationships have never been evaluated in an experimental system. Methods: To test whether hematopoietic cell mutations in PPM1D can increase the susceptibility to cardiac stress, we evaluated cardiac dysfunction in response to angiotensin II infusion in a mouse model where clonal-hematopoiesis-associated mutations in Ppm1d were produced by CRISPR-Cas9 technology. Results: Mice transplanted with hematopoietic stem cells containing clinically relevant mutations in exon 6 of Ppm1d exhibited augmented cardiac remodeling following the continuous infusion of angiotensin II. Ppm1d -mutated macrophages showed impairments in the DDR pathway and had an augmented proinflammatory profile. Mice transplanted with Ppm1d mutated cells exhibited elevated IL-1β in the stressed myocardium, and bone marrow derived macrophages produced more IL-1β in response to LPS stimulation. The administration of an NLRP3 inflammasome inhibitor to mice reversed the cardiac phenotype induced by the Ppm1d -mutated hematopoietic stem cells under conditions of Angiotensin II-induced stress. Conclusions: A mouse model of Ppm1d -mediated clonal hematopoiesis was more susceptible to cardiac stress following of angiotensin II infusion. Mechanistically, disruption of the DDR pathway led to elevations in inflammatory cytokine production, and the NLRP3 inflammasome was shown to be essential for this augmented cardiac stress response. These data indicate that therapy-related clonal hematopoiesis involving mutations in PPM1D could contribute to the cardiac dysfunction observed in cancer survivors.


Leukemia ◽  
2019 ◽  
Vol 33 (11) ◽  
pp. 2732-2766
Author(s):  
Kohei Hosokawa ◽  
Hiroki Mizumaki ◽  
Mahmoud I. Elbadry ◽  
Chizuru Saito ◽  
J. Luis Espinoza ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 40-40
Author(s):  
Maria M. Aivalioti ◽  
Tushar D Bhagat ◽  
Aditi Paranjpe ◽  
Boris Bartholdy ◽  
Kith Pradhan ◽  
...  

Acute myeloid leukemia (AML) is the most frequent leukemia in elderly individuals with a median age at diagnosis of 67 years (Juliusson et al., Blood 2009). It arises in a step-wise process and originates from hematopoietic stem cells (HSC) (Jan et al.,Sci Transl Med. 2012). Genetic and epigenetic alterations drive the formation of pre-leukemic HSC clones with altered function, which can gain dominance and eventually give rise to AML upon the acquisition of cooperating lesions (Jan et al.,Sci Transl Med. 2012). However, it is currently impossible to predict which healthy elderly individuals with clonal hematopoiesis will eventually develop myeloid malignancies, as the pathways to leukemia are unknown. Heterozygous inactivating mutations of the epigenetic regulator Ten-Eleven Translocation-2 (TET2) are commonly found in patients with AML, yet also in a remarkable fraction of healthy elderly individuals in whom it is associated with clonal hematopoiesis (Busque, et al Nat Genet. 2012). These observations and studies in Tet2-deficient mice strongly suggest that TET2 inactivation is an early event in the pathogenesis of myeloid malignancies, but is not sufficient to fully transform HSC (Moran-Crusio et al., Cancel Cell 2011). TET2 cooperates with several transcription factors to regulate hematopoiesis (Rasmussen et al., Genome Res 2019), one of which is PU.1 (de la Rica et al., Genome Biol. 2013), an essential transcription factor governing normal hematopoiesis (Iwasaki et al., Blood 2005). In humans, PU.1 activity or expression is only moderately impaired in the majority of AML patients, and remarkably, also in aged HSC (Will et al., Nat Med. 2015), underscoring the essentiality of PU.1. Importantly, PU.1 target genes are frequently found hypermethylated in AML (Sonnet et al., Genome Med. 2014, Kaasinen et al., Nat Commun. 2019), suggesting a profound epigenetic inactivation of the PU.1 network. We hypothesized that moderate impairment of PU.1 abundance, as found in AML, can cooperate with loss-of-function mutations of Tet2 to initiate malignancy. We developed a novel tissue-specific compound mutant mouse model carrying heterozygous deletion of an upstream regulatory element (URE) of Pu.1 along with Tet2 deletion (Vav-iCre+ PU.1URΕ∆/+Tet2+/flox; Vav-iCre+ PU.1URΕ∆/+Tet2flox/flox). While none of the single mutant mice developed AML, compound mutant mice developed aggressive myeloid leukemia whose penetrance and latency exhibited Tet2 dose dependency. The disease presented with leukocytosis, anemia and splenomegaly. By cell morphology analysis of the peripheral blood, bone marrow and spleen, the leukemic mice exhibited accumulation of differentiation-blocked myeloblasts, myelocytes and/or metamyelocytes, that was confirmed using detailed myeloid differentiation markers, distinguishing the disease in immature or mature AML. Furthermore, gold standard in vitro and in vivo assays, assessing both self-renewal and differentiation capacity of double mutant mice-derived cells, revealed that the expanded differentiation-blocked stem and progenitor cells bear aberrant self-renewal and disease-initiating capacities. Comprehensive molecular profiling by next generation sequencing of disease-initiating cells uncovered a substantial overlap with human AML, such as functional GF1b loss with concomitant overexpression of CD90/Thy1 (Thivakaran et al., Haematologica 2018). Importantly, our analyses also revealed transcriptional dysregulation, hypermethylation of PU.1 regulated enhancers with concomitant loss of enhancer activity and alterations in chromatin accessibility of particularly genes co-bound by PU.1 and TET2. Current efforts focus on identifying key effectors of the dysregulated PU.1/TET2 sub-network driving malignant transformation in clonal hematopoiesis. Our collected data provide proof of concept that moderate PU.1 dose impairment can functionally cooperate with the inactivation of Tet2 in the initiation of myeloid leukemia and uncovers a likely unifying AML pathomechansim. Disclosures Will: Novartis Pharmaceuticals: Other: Service on advisory boards, Research Funding.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Takeshi Fujino ◽  
Susumu Goyama ◽  
Yuki Sugiura ◽  
Daichi Inoue ◽  
Shuhei Asada ◽  
...  

AbstractSomatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3274-3274
Author(s):  
Tre D. Artis ◽  
Vijay G. Sankaran ◽  
Alexander G. Bick

Abstract Clonal hematopoiesis (CH) is the age-related expansion of hematopoietic stem and progenitor cells (HSPCs) due to acquired genetic changes and is associated with increased blood cancer risk. Despite considerable progress in understanding how specific acquired somatic mutations result in clonal expansion, we have a limited understanding of the role of germline mutations that also predispose to clonal expansion. Recent work has revealed a low frequency germline variant found exclusively in individuals of African diasporic descent that confers a 2.4-fold increased risk for CH (Bick et al., Nature, 2020). Remarkably, this variant is found within a putative enhancer of the TET2 gene, which encodes a key epigenetic modifier that catalyzes conversion of methylated cytosines to 5-hydroxymethylcytosine, thereby facilitating DNA demethylation. However, the precise role of this enhancer variant in altering TET2 activity and human hematopoiesis is poorly understood. We specifically hypothesize that this germline variant may alter TET2 activity subtly in hematopoietic stem cells (HSCs) to modify DNA methylation at a number of HSC regulatory elements and subsequently gene expression, which are likely mediated through selective changes in transcription factor (TF) activity. To explore this hypothesis, we performed deletions of the putative TET2 enhancer in adult CD34 +HSPCs using CRISPR/Cas9 editing through the introduction of Cas9 ribonucleoproteins. Following deletion of this enhancer, we observed a moderate increase in the total number of phenotypic long-term reconstituting hematopoietic stem cells (LT-HSCs; as marked by CD34 +CD45RA -CD90 +CD201 +CD133 +CD49c +) and in primary colony formation unit (CFU) assays compared to control editing (AAVS1 edited). Interestingly, the enhancer deletion did not cause an increase in the number of phenotypic short-term HSCs (CD34 +CD45RA -CD90 +CD201 +CD133 +CD49c -), HSPC proliferation, or secondary CFU plating. Assessment of the deletion stability showed selection against enhancer deleted cells during myeloid differentiation, however some cells could still be identified after more than 30 days following editing. This result suggests that the enhancer likely functions in a selective manner within HSCs. The overall phenotypes we observe suggest some overlap, but distinct presentations in comparison to concomitant TET2 coding disruption that we have also performed. Ongoing studies seek to use these promising phenotypic results to define changes in accessible chromatin and DNA methylation states in this isogenic perturbed model. This will enable insights into the specific enhancers altered through this perturbation and we plan to examine TF motif alterations at these regulatory elements. While the perturbation performed may represent a larger perturbation than is seen through the naturally-occurring variant, this provides a platform for defining how this CH predisposition arises with a larger impact perturbation. Use of base editors in a similar manner should enable more selective perturbations, as well. Together, these results will further reveal potential germline genetic and molecular origins of CH and further explain broader mechanisms of TET2 function and the importance of proper DNA methylation during human hematopoiesis that may provide clinical relevance for the potential prevention of blood cancers. Disclosures Sankaran: Cellarity: Consultancy; Forma: Consultancy; Novartis: Consultancy; Ensoma: Consultancy; Branch Biosciences: Consultancy.


2017 ◽  
Vol 21 (4) ◽  
pp. 547-555.e8 ◽  
Author(s):  
Zuzana Tothova ◽  
John M. Krill-Burger ◽  
Katerina D. Popova ◽  
Catherine C. Landers ◽  
Quinlan L. Sievers ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4064-4064
Author(s):  
Sean L. O’Connor ◽  
Carmen A. Sepulveda ◽  
Indreshpal Kaur ◽  
Robert D. Sumari ◽  
John D. McMannis

Abstract Numerous cardiology groups are actively pursuing the use of hematopoietic stem cells for myocardial delivery. Most published studies have used a manual ficoll density separation procedure to enrich the Mononuclear Cell (MNC) population thus enriching the stem cell content in the final therapeutic cell product. Manual ficoll procedures have certain intrinsic limitations including technologist variability, training time, and risk of contamination due to its open nature. Most open and manual cell processing procedures have been supplanted by recent technology developments in automated and closed devices. The BioSafe SEPAX is a closed system-automated ficoll density separation device that currently has FDA 510k clearance for the processing of umbilical cord blood products. In support of multi-center clinical studies being conducted by the NHLBI Cardiovascular Cell Therapy Research Network (CCTRN), the MD Anderson Cell Therapy Laboratory has evaluated bone marrow derived, MNC enriched, stem cells for their cardiovascular therapeutic potential. Traditional transplantation immunophenotyping was performed on these cells to evaluate hematopoietic stem cells (CD34+ and CD133+), T-Cell subsets, B-Cells, and NK-Cells. Colony Forming Units (CFU) as wells as surrogates used in cardiac regeneration studies including endothelial invasion assays and CXCR4 immunophenotyping (a chemokine receptor involved in cell migration) were also performed. Our data suggests the cells produced using the automated ficoll BioSafe SEPAX device are equivalent to cells produced using a manual ficoll procedure with respect to TNC, MNC, viability, and CD34+ recoveries. Cell subsets and function including invasion and CXCR4 analysis were also similar between the two procedures. The automated method produced a product with statistically higher CFU potential than the manual method suggesting the SEPAX isolates a more potent subset of the mononuclear fraction. These studies support the use of the SEPAX device as for enriching BM-MNCs for the CCTRN. Additional benefits might include the use of a closed system, ease of training, speed of cell manufacturing, and product consistency.


2006 ◽  
Author(s):  
Hideyo Hirai ◽  
Pu Zhang ◽  
Tajhal Dayaram ◽  
Christopher Hetherington ◽  
Shin-ichi Mizuno ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document