30 results on '"Chaligne R"'
Search Results
2. Lineage plasticity in prostate cancer depends on FGFR and JAK/STAT inflammatory signaling
- Author
-
Chan, J., primary, Zaidi, S., additional, Love, J., additional, Zhao, J., additional, Setty, M., additional, Wadosky, K., additional, Gopalan, A., additional, Choo, Z.N., additional, Persad, S., additional, Chaudhary, O., additional, Xu, T., additional, Masilionis, I., additional, Morris, M., additional, Mazutis, L., additional, Chaligne, R., additional, Chen, Y., additional, Goodrich, D., additional, Karthaus, W., additional, Pe’er, D., additional, and Sawyers, C., additional
- Published
- 2022
- Full Text
- View/download PDF
3. 5 Oral - Lineage plasticity in prostate cancer depends on FGFR and JAK/STAT inflammatory signaling
- Author
-
Chan, J., Zaidi, S., Love, J., Zhao, J., Setty, M., Wadosky, K., Gopalan, A., Choo, Z.N., Persad, S., Chaudhary, O., Xu, T., Masilionis, I., Morris, M., Mazutis, L., Chaligne, R., Chen, Y., Goodrich, D., Karthaus, W., Pe’er, D., and Sawyers, C.
- Published
- 2022
- Full Text
- View/download PDF
4. Expression illégitime du récepteur de GIP dans les adénomes somatotropes chez les patients acromégales avec réponse paradoxale de la GH à l’hyperglycémie
- Author
-
Hage, M., primary, Viengchareun, S., additional, Chaligne, R., additional, Tosca, L., additional, Adam, C., additional, Parker, F., additional, Villa, C., additional, Lombes, M., additional, Gaillard, S., additional, Chanson, P., additional, and Kamenicky, P., additional
- Published
- 2017
- Full Text
- View/download PDF
5. Expression surrénalienne du glucose-dependent insulinotropic peptide receptor (GIPR) et microduplications de la région 19q13 dans le syndrome de Cushing dépendant de l’alimentation
- Author
-
Lecoq, A.L., Stratakis, C., Viengchareun, S., Chaligné, R., Tosca, L., Deméocq, V., Hage, M., Berthon, A., Hanna, P., Young, J., Lombès, M., Bourdeau, I., Maiter, D., Tabarin, A., Bertherat, J., Lefebvre, H., De Herder, W., Louiset, E., Lacroix, A., Chanson, P., Bouligand, J., and Kamenicky, P.
- Published
- 2017
- Full Text
- View/download PDF
6. Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse
- Author
-
DeWolf, S, primary, Elhanati, Y, additional, Nichols, K, additional, Waters, NR, additional, Nguyen, CL, additional, Slingerland, JB, additional, Rodriguez, N, additional, Lyudovyk, O, additional, Giardina, PA, additional, Kousa, AI, additional, Andrlová, H, additional, Ceglia, N, additional, Fe, T, additional, Kappagantual, R, additional, Li, Y, additional, Aleynick, N, additional, Baez, P, additional, Murali, R, additional, Hayashi, A, additional, Lee, N, additional, Gipson, B, additional, Rangesa, M, additional, Katsamakis, Z, additional, Dai, A, additional, Blouin, AG, additional, Arcila, M, additional, Masilionis, I, additional, Chaligne, R, additional, Ponce, DM, additional, Landau, HJ, additional, Politikos, I, additional, Tamari, R, additional, Hanash, AM, additional, Jenq, RR, additional, Giralt, SA, additional, Markey, KA, additional, Zhang, Y, additional, Perales, M, additional, Socci, ND, additional, Greenbaum, BD, additional, Iacobuzio-Donahue, CA, additional, Hollmann, TJ, additional, van den Brink, MRM, additional, and Peled, U, additional
- Full Text
- View/download PDF
7. Ordered chromatin changes and human X chromosome reactivation by cell fusion-mediated pluripotent reprogramming
- Author
-
Cantone, Irene, Bagci, Hakan, Dormann, Dirk, Dharmalingam, Gopuraja, Nesterova, Tatyana, Brockdorff, Neil, Rougeulle, Claire, Vallot, Celine, Heard, Edith, Chaligne, Ronan, Merkenschlager, Matthias, Fisher, Amanda G., Centre épigénétique et destin cellulaire (EDC (UMR_7216)), Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Cantone, I., Bagci, H., Dormann, D., Dharmalingam, G., Nesterova, T., Brockdorff, N., Rougeulle, C., Vallot, C., Heard, E., Chaligne, R., Merkenschlager, M., and Fisher, A. G.
- Subjects
Cell Nucleus ,Male ,Chromosomes, Human, X ,Science ,[SDV]Life Sciences [q-bio] ,Induced Pluripotent Stem Cells ,Mitosis ,Mouse Embryonic Stem Cells ,Fibroblasts ,Cellular Reprogramming ,Article ,Chromatin ,Cell Line ,Epigenesis, Genetic ,Cell Fusion ,Histones ,Mice ,X Chromosome Inactivation ,Animals ,Humans ,Female ,RNA, Long Noncoding ,ComputingMilieux_MISCELLANEOUS - Abstract
Erasure of epigenetic memory is required to convert somatic cells towards pluripotency. Reactivation of the inactive X chromosome (Xi) has been used to model epigenetic reprogramming in mouse, but human studies are hampered by Xi epigenetic instability and difficulties in tracking partially reprogrammed iPSCs. Here we use cell fusion to examine the earliest events in the reprogramming-induced Xi reactivation of human female fibroblasts. We show that a rapid and widespread loss of Xi-associated H3K27me3 and XIST occurs in fused cells and precedes the bi-allelic expression of selected Xi-genes by many heterokaryons (30–50%). After cell division, RNA-FISH and RNA-seq analyses confirm that Xi reactivation remains partial and that induction of human pluripotency-specific XACT transcripts is rare (1%). These data effectively separate pre- and post-mitotic events in reprogramming-induced Xi reactivation and reveal a complex hierarchy of epigenetic changes that are required to reactivate the genes on the human Xi chromosome., Reactivation of the inactive X chromosome (Xi) has modelled epigenetic reprogramming in mouse. Here, by using cell fusion between human female fibroblasts and mouse embryonic stem cells, the authors show a complex hierarchy of epigenetic changes that are required to reactivate the genes on the human Xi chromosome.
- Published
- 2016
8. RAS-mutant leukaemia stem cells drive clinical resistance to venetoclax.
- Author
-
Sango J, Carcamo S, Sirenko M, Maiti A, Mansour H, Ulukaya G, Tomalin LE, Cruz-Rodriguez N, Wang T, Olszewska M, Olivier E, Jaud M, Nadorp B, Kroger B, Hu F, Silverman L, Chung SS, Wagenblast E, Chaligne R, Eisfeld AK, Demircioglu D, Landau DA, Lito P, Papaemmanuil E, DiNardo CD, Hasson D, Konopleva M, and Papapetrou EP
- Abstract
Cancer driver mutations often show distinct temporal acquisition patterns, but the biological basis for this, if any, remains unknown. RAS mutations occur invariably late in the course of acute myeloid leukaemia, upon progression or relapsed/refractory disease
1-6 . Here, by using human leukaemogenesis models, we first show that RAS mutations are obligatory late events that need to succeed earlier cooperating mutations. We provide the mechanistic explanation for this in a requirement for mutant RAS to specifically transform committed progenitors of the myelomonocytic lineage (granulocyte-monocyte progenitors) harbouring previously acquired driver mutations, showing that advanced leukaemic clones can originate from a different cell type in the haematopoietic hierarchy than ancestral clones. Furthermore, we demonstrate that RAS-mutant leukaemia stem cells (LSCs) give rise to monocytic disease, as observed frequently in patients with poor responses to treatment with the BCL2 inhibitor venetoclax. We show that this is because RAS-mutant LSCs, in contrast to RAS-wild-type LSCs, have altered BCL2 family gene expression and are resistant to venetoclax, driving clinical resistance and relapse with monocytic features. Our findings demonstrate that a specific genetic driver shapes the non-genetic cellular hierarchy of acute myeloid leukaemia by imposing a specific LSC target cell restriction and critically affects therapeutic outcomes in patients., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
9. Engineering mtDNA Deletions by Reconstituting End-Joining in Human Mitochondria.
- Author
-
Fu Y, Land M, Cui R, Kavlashvili T, Kim M, Lieber T, Ryu KW, DeBitetto E, Masilionis I, Saha R, Takizawa M, Baker D, Tigano M, Reznik E, Sharma R, Chaligne R, Thompson CB, Pe'er D, and Sfeir A
- Abstract
Recent breakthroughs in the genetic manipulation of mitochondrial DNA (mtDNA) have enabled the precise introduction of base substitutions and the effective removal of genomes carrying harmful mutations. However, the reconstitution of mtDNA deletions responsible for severe mitochondrial myopathies and age-related diseases has not yet been achieved in human cells. Here, we developed a method to engineer specific mtDNA deletions in human cells by co-expressing end-joining (EJ) machinery and targeted endonucleases. As a proof-of-concept, we used mito-EJ and mito-ScaI to generate a panel of clonal cell lines harboring a ∼3.5 kb mtDNA deletion with the full spectrum of heteroplasmy. Investigating these isogenic cells revealed a critical threshold of ∼75% deleted genomes, beyond which cells exhibited depletion of OXPHOS proteins, severe metabolic disruption, and impaired growth in galactose-containing media. Single-cell multiomic analysis revealed two distinct patterns of nuclear gene deregulation in response to mtDNA deletion accumulation; one triggered at the deletion threshold and another progressively responding to increasing heteroplasmy. In summary, the co-expression of mito-EJ and programable nucleases provides a powerful tool to model disease-associated mtDNA deletions in different cell types. Establishing a panel of cell lines with a large-scale deletion at varying levels of heteroplasmy is a valuable resource for understanding the impact of mtDNA deletions on diseases and guiding the development of potential therapeutic strategies., Highlights: Combining prokaryotic end-joining with targeted endonucleases generates specific mtDNA deletions in human cellsEngineering a panel of cell lines with a large-scale deletion that spans the full spectrum of heteroplasmy75% heteroplasmy is the threshold that triggers mitochondrial and cellular dysfunctionTwo distinct nuclear transcriptional programs in response to mtDNA deletions: threshold-triggered and heteroplasmy-sensing.
- Published
- 2024
- Full Text
- View/download PDF
10. Aging limits stemness and tumorigenesis in the lung by reprogramming iron homeostasis.
- Author
-
Zhuang X, Wang Q, Joost S, Ferrena A, Humphreys DT, Li Z, Blum M, Bastl K, Ding S, Landais Y, Zhan Y, Zhao Y, Chaligne R, Lee JH, Carrasco SE, Bhanot UK, Koche RP, Bott MJ, Katajisto P, Soto-Feliciano YM, Pisanic T, Thomas T, Zheng D, Wong ES, and Tammela T
- Abstract
Aging is associated with a decline in the number and fitness of adult stem cells
1-4 . Aging-associated loss of stemness is posited to suppress tumorigenesis5,6 , but this hypothesis has not been tested in vivo . Here, using physiologically aged autochthonous genetically engineered mouse models and primary cells7,8 , we demonstrate aging suppresses lung cancer initiation and progression by degrading stemness of the alveolar cell of origin. This phenotype is underpinned by aging-associated induction of the transcription factor NUPR1 and its downstream target lipocalin-2 in the cell of origin in mice and humans, leading to a functional iron insufficiency in the aged cells. Genetic inactivation of the NUPR1-lipocalin-2 axis or iron supplementation rescue stemness and promote tumorigenic potential of aged alveolar cells. Conversely, targeting the NUPR1- lipocalin-2 axis is detrimental to young alveolar cells via induction of ferroptosis. We find that aging-associated DNA hypomethylation at specific enhancer sites associates with elevated NUPR1 expression, which is recapitulated in young alveolar cells by inhibition of DNA methylation. We uncover that aging drives a functional iron insufficiency, which leads to loss of stemness and tumorigenesis, but promotes resistance to ferroptosis. These findings have significant implications for the therapeutic modulation of cellular iron homeostasis in regenerative medicine and in cancer prevention. Furthermore, our findings are consistent with a model whereby most human cancers initiate in young individuals, revealing a critical window for such cancer prevention efforts.- Published
- 2024
- Full Text
- View/download PDF
11. The covariance environment defines cellular niches for spatial inference.
- Author
-
Haviv D, Remšík J, Gatie M, Snopkowski C, Takizawa M, Pereira N, Bashkin J, Jovanovich S, Nawy T, Chaligne R, Boire A, Hadjantonakis AK, and Pe'er D
- Abstract
A key challenge of analyzing data from high-resolution spatial profiling technologies is to suitably represent the features of cellular neighborhoods or niches. Here we introduce the covariance environment (COVET), a representation that leverages the gene-gene covariate structure across cells in the niche to capture the multivariate nature of cellular interactions within it. We define a principled optimal transport-based distance metric between COVET niches that scales to millions of cells. Using COVET to encode spatial context, we developed environmental variational inference (ENVI), a conditional variational autoencoder that jointly embeds spatial and single-cell RNA sequencing data into a latent space. ENVI includes two decoders: one to impute gene expression across the spatial modality and a second to project spatial information onto single-cell data. ENVI can confer spatial context to genomics data from single dissociated cells and outperforms alternatives for imputing gene expression on diverse spatial datasets., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Childhood cancer mutagenesis caused by transposase-derived PGBD5.
- Author
-
Yamada M, Keller RR, Gutierrez RL, Cameron D, Suzuki H, Sanghrajka R, Vaynshteyn J, Gerwin J, Maura F, Hooper W, Shah M, Robine N, Demarest P, Bayin NS, Zapater LJ, Reed C, Hébert S, Masilionis I, Chaligne R, Socci ND, Taylor MD, Kleinman CL, Joyner AL, Raju GP, and Kentsis A
- Subjects
- Humans, Child, Animals, Mice, Transposases genetics, Transposases metabolism, Hedgehog Proteins metabolism, Transcription Factors genetics, Mutagenesis, Medulloblastoma genetics, Cerebellar Neoplasms genetics
- Abstract
Genomic rearrangements are a hallmark of most childhood tumors, including medulloblastoma, one of the most common brain tumors in children, but their causes remain largely unknown. Here, we show that PiggyBac transposable element derived 5 (Pgbd5) promotes tumor development in multiple developmentally accurate mouse models of Sonic Hedgehog (SHH) medulloblastoma. Most Pgbd5-deficient mice do not develop tumors, while maintaining normal cerebellar development. Ectopic activation of SHH signaling is sufficient to enforce cerebellar granule cell progenitor-like cell states, which exhibit Pgbd5-dependent expression of distinct DNA repair and neurodevelopmental factors. Mouse medulloblastomas expressing Pgbd5 have increased numbers of somatic structural DNA rearrangements, some of which carry PGBD5-specific sequences at their breakpoints. Similar sequence breakpoints recurrently affect somatic DNA rearrangements of known tumor suppressors and oncogenes in medulloblastomas in 329 children. This identifies PGBD5 as a medulloblastoma mutator and provides a genetic mechanism for the generation of oncogenic DNA rearrangements in childhood cancer.
- Published
- 2024
- Full Text
- View/download PDF
13. Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse.
- Author
-
DeWolf S, Elhanati Y, Nichols K, Waters NR, Nguyen CL, Slingerland JB, Rodriguez N, Lyudovyk O, Giardina PA, Kousa AI, Andrlová H, Ceglia N, Fei T, Kappagantula R, Li Y, Aleynick N, Baez P, Murali R, Hayashi A, Lee N, Gipson B, Rangesa M, Katsamakis Z, Dai A, Blouin AG, Arcila M, Masilionis I, Chaligne R, Ponce DM, Landau HJ, Politikos I, Tamari R, Hanash AM, Jenq RR, Giralt SA, Markey KA, Zhang Y, Perales MA, Socci ND, Greenbaum BD, Iacobuzio-Donahue CA, Hollmann TJ, van den Brink MRM, and Peled JU
- Subjects
- Humans, Mice, Animals, T-Lymphocytes pathology, Receptors, Antigen, T-Cell, Hematopoietic Stem Cell Transplantation, Graft vs Host Disease pathology
- Abstract
T cells are the central drivers of many inflammatory diseases, but the repertoire of tissue-resident T cells at sites of pathology in human organs remains poorly understood. We examined the site-specificity of T cell receptor (TCR) repertoires across tissues (5 to 18 tissues per patient) in prospectively collected autopsies of patients with and without graft-versus-host disease (GVHD), a potentially lethal tissue-targeting complication of allogeneic hematopoietic cell transplantation, and in mouse models of GVHD. Anatomic similarity between tissues was a key determinant of TCR repertoire composition within patients, independent of disease or transplant status. The T cells recovered from peripheral blood and spleens in patients and mice captured a limited portion of the TCR repertoire detected in tissues. Whereas few T cell clones were shared across patients, motif-based clustering revealed shared repertoire signatures across patients in a tissue-specific fashion. T cells at disease sites had a tissue-resident phenotype and were of donor origin based on single-cell chimerism analysis. These data demonstrate the complex composition of T cell populations that persist in human tissues at the end stage of an inflammatory disorder after lymphocyte-directed therapy. These findings also underscore the importance of studying T cell in tissues rather than blood for tissue-based pathologies and suggest the tissue-specific nature of both the endogenous and posttransplant T cell landscape.
- Published
- 2023
- Full Text
- View/download PDF
14. MACHETE identifies interferon-encompassing chromosome 9p21.3 deletions as mediators of immune evasion and metastasis.
- Author
-
Barriga FM, Tsanov KM, Ho YJ, Sohail N, Zhang A, Baslan T, Wuest AN, Del Priore I, Meškauskaitė B, Livshits G, Alonso-Curbelo D, Simon J, Chaves-Perez A, Bar-Sagi D, Iacobuzio-Donahue CA, Notta F, Chaligne R, Sharma R, Pe'er D, and Lowe SW
- Subjects
- Animals, Mice, Chromosome Deletion, Chromosomes, Immune Evasion, Tumor Microenvironment genetics, Tandem Repeat Sequences, Interferons, Neoplasms
- Abstract
The most prominent homozygous deletions in cancer affect chromosome 9p21.3 and eliminate CDKN2A/B tumor suppressors, disabling a cell-intrinsic barrier to tumorigenesis. Half of 9p21.3 deletions, however, also encompass a type I interferon (IFN) gene cluster; the consequences of this co-deletion remain unexplored. To functionally dissect 9p21.3 and other large genomic deletions, we developed a flexible deletion engineering strategy, MACHETE (molecular alteration of chromosomes with engineered tandem elements). Applying MACHETE to a syngeneic mouse model of pancreatic cancer, we found that co-deletion of the IFN cluster promoted immune evasion, metastasis and immunotherapy resistance. Mechanistically, IFN co-deletion disrupted type I IFN signaling in the tumor microenvironment, leading to marked changes in infiltrating immune cells and escape from CD8
+ T-cell surveillance, effects largely driven by the poorly understood interferon epsilon. These results reveal a chromosomal deletion that disables both cell-intrinsic and cell-extrinsic tumor suppression and provide a framework for interrogating large deletions in cancer and beyond., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
15. Systematic Comparison of Pancreatic Ductal Adenocarcinoma Models Identifies a Conserved Highly Plastic Basal Cell State.
- Author
-
Pitter KL, Grbovic-Huezo O, Joost S, Singhal A, Blum M, Wu K, Holm M, Ferrena A, Bhutkar A, Hudson A, Lecomte N, de Stanchina E, Chaligne R, Iacobuzio-Donahue CA, Pe'er D, and Tammela T
- Subjects
- Humans, Pancreatic Ducts, Plastics, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Intratumoral heterogeneity and cellular plasticity have emerged as hallmarks of cancer, including pancreatic ductal adenocarcinoma (PDAC). As PDAC portends a dire prognosis, a better understanding of the mechanisms underpinning cellular diversity in PDAC is crucial. Here, we investigated the cellular heterogeneity of PDAC cancer cells across a range of in vitro and in vivo growth conditions using single-cell genomics. Heterogeneity contracted significantly in two-dimensional and three-dimensional cell culture models but was restored upon orthotopic transplantation. Orthotopic transplants reproducibly acquired cell states identified in autochthonous PDAC tumors, including a basal state exhibiting coexpression and coaccessibility of epithelial and mesenchymal genes. Lineage tracing combined with single-cell transcriptomics revealed that basal cells display high plasticity in situ. This work defines the impact of cellular growth conditions on phenotypic diversity and uncovers a highly plastic cell state with the capacity to facilitate state transitions and promote intratumoral heterogeneity in PDAC., Significance: This work provides important insights into how different model systems of pancreatic ductal adenocarcinoma mold the phenotypic space of cancer cells, highlighting the power of in vivo models., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
16. Single-cell multi-omics of human clonal hematopoiesis reveals that DNMT3A R882 mutations perturb early progenitor states through selective hypomethylation.
- Author
-
Nam AS, Dusaj N, Izzo F, Murali R, Myers RM, Mouhieddine TH, Sotelo J, Benbarche S, Waarts M, Gaiti F, Tahri S, Levine R, Abdel-Wahab O, Godley LA, Chaligne R, Ghobrial I, and Landau DA
- Subjects
- DNA Modification Methylases genetics, Hematopoiesis genetics, Humans, Mutation, Polycomb Repressive Complex 2 genetics, Clonal Hematopoiesis, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A genetics
- Abstract
Somatic mutations in cancer genes have been detected in clonal expansions across healthy human tissue, including in clonal hematopoiesis. However, because mutated and wild-type cells are admixed, we have limited ability to link genotypes with phenotypes. To overcome this limitation, we leveraged multi-modality single-cell sequencing, capturing genotype, transcriptomes and methylomes in progenitors from individuals with DNMT3A R882 mutated clonal hematopoiesis. DNMT3A mutations result in myeloid over lymphoid bias, and an expansion of immature myeloid progenitors primed toward megakaryocytic-erythroid fate, with dysregulated expression of lineage and leukemia stem cell markers. Mutated DNMT3A leads to preferential hypomethylation of polycomb repressive complex 2 targets and a specific CpG flanking motif. Notably, the hypomethylation motif is enriched in binding motifs of key hematopoietic transcription factors, serving as a potential mechanistic link between DNMT3A mutations and aberrant transcriptional phenotypes. Thus, single-cell multi-omics paves the road to defining the downstream consequences of mutations that drive clonal mosaicism., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
17. Lineage plasticity in prostate cancer depends on JAK/STAT inflammatory signaling.
- Author
-
Chan JM, Zaidi S, Love JR, Zhao JL, Setty M, Wadosky KM, Gopalan A, Choo ZN, Persad S, Choi J, LaClair J, Lawrence KE, Chaudhary O, Xu T, Masilionis I, Linkov I, Wang S, Lee C, Barlas A, Morris MJ, Mazutis L, Chaligne R, Chen Y, Goodrich DW, Karthaus WR, Pe'er D, and Sawyers CL
- Subjects
- Androgen Antagonists, Animals, Humans, Janus Kinase Inhibitors therapeutic use, Male, Mice, Neoplasms, Experimental, Organoids, Signal Transduction, Cell Plasticity, Drug Resistance, Neoplasm, ErbB Receptors, Janus Kinases genetics, Janus Kinases metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, STAT Transcription Factors genetics, STAT Transcription Factors metabolism
- Abstract
Drug resistance in cancer is often linked to changes in tumor cell state or lineage, but the molecular mechanisms driving this plasticity remain unclear. Using murine organoid and genetically engineered mouse models, we investigated the causes of lineage plasticity in prostate cancer and its relationship to antiandrogen resistance. We found that plasticity initiates in an epithelial population defined by mixed luminal-basal phenotype and that it depends on increased Janus kinase (JAK) and fibroblast growth factor receptor (FGFR) activity. Organoid cultures from patients with castration-resistant disease harboring mixed-lineage cells reproduce the dependency observed in mice by up-regulating luminal gene expression upon JAK and FGFR inhibitor treatment. Single-cell analysis confirms the presence of mixed-lineage cells with increased JAK/STAT (signal transducer and activator of transcription) and FGFR signaling in a subset of patients with metastatic disease, with implications for stratifying patients for clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
18. MAIT and Vδ2 unconventional T cells are supported by a diverse intestinal microbiome and correlate with favorable patient outcome after allogeneic HCT.
- Author
-
Andrlová H, Miltiadous O, Kousa AI, Dai A, DeWolf S, Violante S, Park HY, Janaki-Raman S, Gardner R, El Daker S, Slingerland J, Giardina P, Clurman A, Gomes ALC, Nguyen C, da Silva MB, Armijo GK, Lee N, Zappasodi R, Chaligne R, Masilionis I, Fontana E, Ponce D, Cho C, Bush A, Hill L, Chao N, Sung AD, Giralt S, Vidal EH, Hosszu KK, Devlin SM, Peled JU, Cross JR, Perales MA, Godfrey DI, van den Brink MRM, and Markey KA
- Subjects
- Humans, Ligands, Gastrointestinal Microbiome, Graft vs Host Disease, Hematopoietic Stem Cell Transplantation, Mucosal-Associated Invariant T Cells
- Abstract
Microbial diversity is associated with improved outcomes in recipients of allogeneic hematopoietic cell transplantation (allo-HCT), but the mechanism underlying this observation is unclear. In a cohort of 174 patients who underwent allo-HCT, we demonstrate that a diverse intestinal microbiome early after allo-HCT is associated with an increased number of innate-like mucosal-associated invariant T (MAIT) cells, which are in turn associated with improved overall survival and less acute graft-versus-host disease (aGVHD). Immune profiling of conventional and unconventional immune cell subsets revealed that the prevalence of Vδ2 cells, the major circulating subpopulation of γδ T cells, closely correlated with the frequency of MAIT cells and was associated with less aGVHD. Analysis of these populations using both single-cell transcriptomics and flow cytometry suggested a shift toward activated phenotypes and a gain of cytotoxic and effector functions after transplantation. A diverse intestinal microbiome with the capacity to produce activating ligands for MAIT and Vδ2 cells appeared to be necessary for the maintenance of these populations after allo-HCT. These data suggest an immunological link between intestinal microbial diversity, microbe-derived ligands, and maintenance of unconventional T cells.
- Published
- 2022
- Full Text
- View/download PDF
19. Epigenetic encoding, heritability and plasticity of glioma transcriptional cell states.
- Author
-
Chaligne R, Gaiti F, Silverbush D, Schiffman JS, Weisman HR, Kluegel L, Gritsch S, Deochand SD, Gonzalez Castro LN, Richman AR, Klughammer J, Biancalani T, Muus C, Sheridan C, Alonso A, Izzo F, Park J, Rozenblatt-Rosen O, Regev A, Suvà ML, and Landau DA
- Subjects
- Cell Line, Tumor, CpG Islands genetics, DNA Copy Number Variations genetics, DNA Methylation genetics, Humans, Isocitrate Dehydrogenase genetics, Phylogeny, Polycomb Repressive Complex 2 metabolism, Promoter Regions, Genetic genetics, Single-Cell Analysis, Transcriptome genetics, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Plasticity genetics, Epigenesis, Genetic, Glioma genetics, Glioma pathology, Inheritance Patterns genetics, Transcription, Genetic
- Abstract
Single-cell RNA sequencing has revealed extensive transcriptional cell state diversity in cancer, often observed independently of genetic heterogeneity, raising the central question of how malignant cell states are encoded epigenetically. To address this, here we performed multiomics single-cell profiling-integrating DNA methylation, transcriptome and genotype within the same cells-of diffuse gliomas, tumors characterized by defined transcriptional cell state diversity. Direct comparison of the epigenetic profiles of distinct cell states revealed key switches for state transitions recapitulating neurodevelopmental trajectories and highlighted dysregulated epigenetic mechanisms underlying gliomagenesis. We further developed a quantitative framework to directly measure cell state heritability and transition dynamics based on high-resolution lineage trees in human samples. We demonstrated heritability of malignant cell states, with key differences in hierarchal and plastic cell state architectures in IDH-mutant glioma versus IDH-wild-type glioblastoma, respectively. This work provides a framework anchoring transcriptional cancer cell states in their epigenetic encoding, inheritance and transition dynamics., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
20. Discovery of Candidate DNA Methylation Cancer Driver Genes.
- Author
-
Pan H, Renaud L, Chaligne R, Bloehdorn J, Tausch E, Mertens D, Fink AM, Fischer K, Zhang C, Betel D, Gnirke A, Imielinski M, Moreaux J, Hallek M, Meissner A, Stilgenbauer S, Wu CJ, Elemento O, and Landau DA
- Subjects
- Epigenesis, Genetic, Humans, DNA Methylation genetics, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
Epigenetic alterations, such as promoter hypermethylation, may drive cancer through tumor suppressor gene inactivation. However, we have limited ability to differentiate driver DNA methylation (DNAme) changes from passenger events. We developed DNAme driver inference-MethSig-accounting for the varying stochastic hypermethylation rate across the genome and between samples. We applied MethSig to bisulfite sequencing data of chronic lymphocytic leukemia (CLL), multiple myeloma, ductal carcinoma in situ , glioblastoma, and to methylation array data across 18 tumor types in TCGA. MethSig resulted in well-calibrated quantile-quantile plots and reproducible inference of likely DNAme drivers with increased sensitivity/specificity compared with benchmarked methods. CRISPR/Cas9 knockout of selected candidate CLL DNAme drivers provided a fitness advantage with and without therapeutic intervention. Notably, DNAme driver risk score was closely associated with adverse outcome in independent CLL cohorts. Collectively, MethSig represents a novel inference framework for DNAme driver discovery to chart the role of aberrant DNAme in cancer. SIGNIFICANCE: MethSig provides a novel statistical framework for the analysis of DNA methylation changes in cancer, to specifically identify candidate DNA methylation driver genes of cancer progression and relapse, empowering the discovery of epigenetic mechanisms that enhance cancer cell fitness. This article is highlighted in the In This Issue feature, p. 2113 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
21. Smart-RRBS for single-cell methylome and transcriptome analysis.
- Author
-
Gu H, Raman AT, Wang X, Gaiti F, Chaligne R, Mohammad AW, Arczewska A, Smith ZD, Landau DA, Aryee MJ, Meissner A, and Gnirke A
- Subjects
- Amino Acid Sequence, Anti-Bacterial Agents pharmacology, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases metabolism, Doxycycline pharmacology, Epigenome, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, Humans, Intracellular Signaling Peptides and Proteins, RNA, Messenger genetics, RNA, Messenger metabolism, Transcriptome, DNA metabolism, Single-Cell Analysis
- Abstract
The integration of DNA methylation and transcriptional state within single cells is of broad interest. Several single-cell dual- and multi-omics approaches have been reported that enable further investigation into cellular heterogeneity, including the discovery and in-depth study of rare cell populations. Such analyses will continue to provide important mechanistic insights into the regulatory consequences of epigenetic modifications. We recently reported a new method for profiling the DNA methylome and transcriptome from the same single cells in a cancer research study. Here, we present details of the protocol and provide guidance on its utility. Our Smart-RRBS (reduced representation bisulfite sequencing) protocol combines Smart-seq2 and RRBS and entails physically separating mRNA from the genomic DNA. It generates paired epigenetic promoter and RNA-expression measurements for ~24% of protein-coding genes in a typical single cell. It also works for micro-dissected tissue samples comprising hundreds of cells. The protocol, excluding flow sorting of cells and sequencing, takes ~3 d to process up to 192 samples manually. It requires basic molecular biology expertise and laboratory equipment, including a PCR workstation with UV sterilization, a DNA fluorometer and a microfluidic electrophoresis system., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
22. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics.
- Author
-
Nam AS, Chaligne R, and Landau DA
- Subjects
- Epigenomics, Humans, Mutation, Single-Cell Analysis, Tumor Microenvironment, Clonal Evolution genetics, Computational Biology, Genetic Variation, Genomics, Neoplasms genetics
- Abstract
Cancer represents an evolutionary process through which growing malignant populations genetically diversify, leading to tumour progression, relapse and resistance to therapy. In addition to genetic diversity, the cell-to-cell variation that fuels evolutionary selection also manifests in cellular states, epigenetic profiles, spatial distributions and interactions with the microenvironment. Therefore, the study of cancer requires the integration of multiple heritable dimensions at the resolution of the single cell - the atomic unit of somatic evolution. In this Review, we discuss emerging analytic and experimental technologies for single-cell multi-omics that enable the capture and integration of multiple data modalities to inform the study of cancer evolution. These data show that cancer results from a complex interplay between genetic and non-genetic determinants of somatic evolution.
- Published
- 2021
- Full Text
- View/download PDF
23. DNA methylation disruption reshapes the hematopoietic differentiation landscape.
- Author
-
Izzo F, Lee SC, Poran A, Chaligne R, Gaiti F, Gross B, Murali RR, Deochand SD, Ang C, Jones PW, Nam AS, Kim KT, Kothen-Hill S, Schulman RC, Ki M, Lhoumaud P, Skok JA, Viny AD, Levine RL, Kenigsberg E, Abdel-Wahab O, and Landau DA
- Subjects
- Animals, DNA (Cytosine-5-)-Methyltransferases genetics, DNA-Binding Proteins genetics, Hematopoietic Stem Cells physiology, Humans, Male, Mice, Mice, Transgenic, Mutation genetics, Transcription, Genetic genetics, Transcriptome genetics, Cell Differentiation genetics, DNA Methylation genetics, Hematopoiesis genetics
- Abstract
Mutations in genes involved in DNA methylation (DNAme; for example, TET2 and DNMT3A) are frequently observed in hematological malignancies
1-3 and clonal hematopoiesis4,5 . Applying single-cell sequencing to murine hematopoietic stem and progenitor cells, we observed that these mutations disrupt hematopoietic differentiation, causing opposite shifts in the frequencies of erythroid versus myelomonocytic progenitors following Tet2 or Dnmt3a loss. Notably, these shifts trace back to transcriptional priming skews in uncommitted hematopoietic stem cells. To reconcile genome-wide DNAme changes with specific erythroid versus myelomonocytic skews, we provide evidence in support of differential sensitivity of transcription factors due to biases in CpG enrichment in their binding motif. Single-cell transcriptomes with targeted genotyping showed similar skews in transcriptional priming of DNMT3A-mutated human clonal hematopoiesis bone marrow progenitors. These data show that DNAme shapes the topography of hematopoietic differentiation, and support a model in which genome-wide methylation changes are transduced to differentiation skews through biases in CpG enrichment of the transcription factor binding motif.- Published
- 2020
- Full Text
- View/download PDF
24. Somatic mutations and cell identity linked by Genotyping of Transcriptomes.
- Author
-
Nam AS, Kim KT, Chaligne R, Izzo F, Ang C, Taylor J, Myers RM, Abu-Zeinah G, Brand R, Omans ND, Alonso A, Sheridan C, Mariani M, Dai X, Harrington E, Pastore A, Cubillos-Ruiz JR, Tam W, Hoffman R, Rabadan R, Scandura JM, Abdel-Wahab O, Smibert P, and Landau DA
- Subjects
- Animals, Antigens, CD34 metabolism, Calreticulin genetics, Cell Line, Cell Proliferation, Clone Cells classification, Clone Cells metabolism, Clone Cells pathology, Endoribonucleases metabolism, Hematopoiesis genetics, Hematopoietic Stem Cells classification, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, High-Throughput Nucleotide Sequencing methods, Humans, Mice, Models, Molecular, Myeloproliferative Disorders classification, NF-kappa B metabolism, Neoplasms classification, Neoplastic Stem Cells cytology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Primary Myelofibrosis genetics, Primary Myelofibrosis pathology, Protein Serine-Threonine Kinases metabolism, Sequence Analysis, RNA methods, Single-Cell Analysis methods, Unfolded Protein Response genetics, Genotype, Mutation, Myeloproliferative Disorders genetics, Myeloproliferative Disorders pathology, Neoplasms genetics, Neoplasms pathology, Transcriptome genetics
- Abstract
Defining the transcriptomic identity of malignant cells is challenging in the absence of surface markers that distinguish cancer clones from one another, or from admixed non-neoplastic cells. To address this challenge, here we developed Genotyping of Transcriptomes (GoT), a method to integrate genotyping with high-throughput droplet-based single-cell RNA sequencing. We apply GoT to profile 38,290 CD34
+ cells from patients with CALR-mutated myeloproliferative neoplasms to study how somatic mutations corrupt the complex process of human haematopoiesis. High-resolution mapping of malignant versus normal haematopoietic progenitors revealed an increasing fitness advantage with myeloid differentiation of cells with mutated CALR. We identified the unfolded protein response as a predominant outcome of CALR mutations, with a considerable dependency on cell identity, as well as upregulation of the NF-κB pathway specifically in uncommitted stem cells. We further extended the GoT toolkit to genotype multiple targets and loci that are distant from transcript ends. Together, these findings reveal that the transcriptional output of somatic mutations in myeloproliferative neoplasms is dependent on the native cell identity.- Published
- 2019
- Full Text
- View/download PDF
25. Epigenetic evolution and lineage histories of chronic lymphocytic leukaemia.
- Author
-
Gaiti F, Chaligne R, Gu H, Brand RM, Kothen-Hill S, Schulman RC, Grigorev K, Risso D, Kim KT, Pastore A, Huang KY, Alonso A, Sheridan C, Omans ND, Biederstedt E, Clement K, Wang L, Felsenfeld JA, Bhavsar EB, Aryee MJ, Allan JN, Furman R, Gnirke A, Wu CJ, Meissner A, and Landau DA
- Subjects
- Base Sequence, Biological Clocks, DNA Methylation, Epigenome genetics, Gene Expression Regulation, Neoplastic, Humans, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Mutation Rate, Sequence Analysis, RNA, Single-Cell Analysis, Transcription, Genetic, Cell Lineage genetics, Epigenesis, Genetic, Evolution, Molecular, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell pathology
- Abstract
Genetic and epigenetic intra-tumoral heterogeneity cooperate to shape the evolutionary course of cancer
1 . Chronic lymphocytic leukaemia (CLL) is a highly informative model for cancer evolution as it undergoes substantial genetic diversification and evolution after therapy2,3 . The CLL epigenome is also an important disease-defining feature4,5 , and growing populations of cells in CLL diversify by stochastic changes in DNA methylation known as epimutations6 . However, previous studies using bulk sequencing methods to analyse the patterns of DNA methylation were unable to determine whether epimutations affect CLL populations homogeneously. Here, to measure the epimutation rate at single-cell resolution, we applied multiplexed single-cell reduced-representation bisulfite sequencing to B cells from healthy donors and patients with CLL. We observed that the common clonal origin of CLL results in a consistently increased epimutation rate, with low variability in the cell-to-cell epimutation rate. By contrast, variable epimutation rates across healthy B cells reflect diverse evolutionary ages across the trajectory of B cell differentiation, consistent with epimutations serving as a molecular clock. Heritable epimutation information allowed us to reconstruct lineages at high-resolution with single-cell data, and to apply this directly to patient samples. The CLL lineage tree shape revealed earlier branching and longer branch lengths than in normal B cells, reflecting rapid drift after the initial malignant transformation and a greater proliferative history. Integration of single-cell bisulfite sequencing analysis with single-cell transcriptomes and genotyping confirmed that genetic subclones mapped to distinct clades, as inferred solely on the basis of epimutation information. Finally, to examine potential lineage biases during therapy, we profiled serial samples during ibrutinib-associated lymphocytosis, and identified clades of cells that were preferentially expelled from the lymph node after treatment, marked by distinct transcriptional profiles. The single-cell integration of genetic, epigenetic and transcriptional information thus charts the lineage history of CLL and its evolution with therapy.- Published
- 2019
- Full Text
- View/download PDF
26. Corrupted coordination of epigenetic modifications leads to diverging chromatin states and transcriptional heterogeneity in CLL.
- Author
-
Pastore A, Gaiti F, Lu SX, Brand RM, Kulm S, Chaligne R, Gu H, Huang KY, Stamenova EK, Béguelin W, Jiang Y, Schulman RC, Kim KT, Alonso A, Allan JN, Furman RR, Gnirke A, Wu CJ, Melnick AM, Meissner A, Bernstein BE, Abdel-Wahab O, and Landau DA
- Subjects
- DNA Methylation, Evolution, Molecular, Gene Silencing, Genes, Immunoglobulin Heavy Chain genetics, Healthy Volunteers, Histone Code genetics, Histones genetics, Histones metabolism, Humans, Leukemia, Lymphocytic, Chronic, B-Cell blood, Polycomb-Group Proteins genetics, Polycomb-Group Proteins metabolism, Promoter Regions, Genetic genetics, Sequence Analysis, RNA, Single-Cell Analysis methods, Exome Sequencing, B-Lymphocytes metabolism, Chromatin metabolism, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
Cancer evolution is fueled by epigenetic as well as genetic diversity. In chronic lymphocytic leukemia (CLL), intra-tumoral DNA methylation (DNAme) heterogeneity empowers evolution. Here, to comprehensively study the epigenetic dimension of cancer evolution, we integrate DNAme analysis with histone modification mapping and single cell analyses of RNA expression and DNAme in 22 primary CLL and 13 healthy donor B lymphocyte samples. Our data reveal corrupted coherence across different layers of the CLL epigenome. This manifests in decreased mutual information across epigenetic modifications and gene expression attributed to cell-to-cell heterogeneity. Disrupted epigenetic-transcriptional coordination in CLL is also reflected in the dysregulation of the transcriptional output as a function of the combinatorial chromatin states, including incomplete Polycomb-mediated gene silencing. Notably, we observe unexpected co-mapping of typically mutually exclusive activating and repressing histone modifications, suggestive of intra-tumoral epigenetic diversity. Thus, CLL epigenetic diversification leads to decreased coordination across layers of epigenetic information, likely reflecting an admixture of cells with diverging cellular identities.
- Published
- 2019
- Full Text
- View/download PDF
27. TARGET-seq Takes Aim at Cancer Evolution through Multi-omics Single-Cell Genotyping and Transcriptomics.
- Author
-
Chaligne R, Nam AS, and Landau DA
- Subjects
- Genotype, Humans, Mutation, Sequence Analysis, RNA, Neoplasms, Single-Cell Analysis
- Abstract
In this issue of Molecular Cell, Rodriguez-Meira et al. (2019) present TARGET-seq, an elegant single-cell method that genotypes somatic mutations and captures whole transcriptomes in the same tumor cells, thus paving the way to directly link somatic mutations with resulting transcriptional phenotypes in clonally diverse cancer populations., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
28. Efficient and versatile CRISPR engineering of human neurons in culture to model neurological disorders.
- Author
-
Shah RR, Cholewa-Waclaw J, Davies FCJ, Paton KM, Chaligne R, Heard E, Abbott CM, and Bird AP
- Abstract
The recent identification of multiple new genetic causes of neurological disorders highlights the need for model systems that give experimental access to the underlying biology. In particular, the ability to couple disease-causing mutations with human neuronal differentiation systems would be beneficial. Gene targeting is a well-known approach for dissecting gene function, but low rates of homologous recombination in somatic cells (including neuronal cells) have traditionally impeded the development of robust cellular models of neurological disorders. Recently, however, CRISPR/Cas9 gene editing technologies have expanded the number of systems within which gene targeting is possible. Here we adopt as a model system LUHMES cells, a commercially available diploid human female mesencephalic cell line that differentiates into homogeneous mature neurons in 1-2 weeks. We describe optimised methods for transfection and selection of neuronal progenitor cells carrying targeted genomic alterations using CRISPR/Cas9 technology. By targeting the endogenous X-linked MECP2 locus, we introduced four independent missense mutations that cause the autism spectrum disorder Rett syndrome and observed the desired genetic structure in 3-26% of selected clones, including gene targeting of the inactive X chromosome. Similar efficiencies were achieved by introducing neurodevelopmental disorder-causing mutations at the autosomal EEF1A2 locus on chromosome 20. Our results indicate that efficiency of genetic "knock-in" is determined by the location of the mutation within the donor DNA molecule. Furthermore, we successfully introduced an mCherry tag at the MECP2 locus to yield a fusion protein, demonstrating that larger insertions are also straightforward in this system. We suggest that our optimised methods for altering the genome of LUHMES cells make them an attractive model for the study of neurogenetic disorders., Competing Interests: No competing interests were disclosed.
- Published
- 2016
- Full Text
- View/download PDF
29. Ordered chromatin changes and human X chromosome reactivation by cell fusion-mediated pluripotent reprogramming.
- Author
-
Cantone I, Bagci H, Dormann D, Dharmalingam G, Nesterova T, Brockdorff N, Rougeulle C, Vallot C, Heard E, Chaligne R, Merkenschlager M, and Fisher AG
- Subjects
- Animals, Cell Fusion methods, Cell Line, Cell Nucleus genetics, Cell Nucleus metabolism, Epigenesis, Genetic, Female, Fibroblasts, Humans, Induced Pluripotent Stem Cells metabolism, Male, Mice, Mitosis, Mouse Embryonic Stem Cells metabolism, Cellular Reprogramming genetics, Chromatin genetics, Chromosomes, Human, X genetics, Histones genetics, RNA, Long Noncoding genetics, X Chromosome Inactivation genetics
- Abstract
Erasure of epigenetic memory is required to convert somatic cells towards pluripotency. Reactivation of the inactive X chromosome (Xi) has been used to model epigenetic reprogramming in mouse, but human studies are hampered by Xi epigenetic instability and difficulties in tracking partially reprogrammed iPSCs. Here we use cell fusion to examine the earliest events in the reprogramming-induced Xi reactivation of human female fibroblasts. We show that a rapid and widespread loss of Xi-associated H3K27me3 and XIST occurs in fused cells and precedes the bi-allelic expression of selected Xi-genes by many heterokaryons (30-50%). After cell division, RNA-FISH and RNA-seq analyses confirm that Xi reactivation remains partial and that induction of human pluripotency-specific XACT transcripts is rare (1%). These data effectively separate pre- and post-mitotic events in reprogramming-induced Xi reactivation and reveal a complex hierarchy of epigenetic changes that are required to reactivate the genes on the human Xi chromosome.
- Published
- 2016
- Full Text
- View/download PDF
30. The hematopoietic stem cell compartment of JAK2V617F-positive myeloproliferative disorders is a reflection of disease heterogeneity.
- Author
-
James C, Mazurier F, Dupont S, Chaligne R, Lamrissi-Garcia I, Tulliez M, Lippert E, Mahon FX, Pasquet JM, Etienne G, Delhommeau F, Giraudier S, Vainchenker W, and de Verneuil H
- Subjects
- Animals, Cell Proliferation, Hematopoietic Stem Cell Transplantation, Humans, Mice, Mice, SCID, Mutation, Missense, Myeloproliferative Disorders genetics, Phenotype, Polycythemia Vera genetics, Polycythemia Vera pathology, Primary Myelofibrosis genetics, Primary Myelofibrosis pathology, Transplantation, Heterologous, Hematopoietic Stem Cells pathology, Janus Kinase 2 genetics, Myeloproliferative Disorders pathology
- Abstract
The JAK2V617F somatic point mutation has been described in patients with myeloproliferative disorders (MPDs). Despite this progress, it remains unknown how a single JAK2 mutation causes 3 different MPD phenotypes, polycythemia vera (PV), essential thrombocythemia, and primitive myelofibrosis (PMF). Using an in vivo xenotransplantation assay in nonobese diabetic-severe combined immunodeficient (NOD/SCID) mice, we tested whether disease heterogeneity was associated with quantitative or qualitative differences in the hematopoietic stem cell (HSC) compartment. We show that the HSC compartment of PV and PMF patients contains JAK2V617F-positive long-term, multipotent, and self-renewing cells. However, the proportion of JAK2V617F and JAK2 wild-type SCID repopulating cells was dramatically different in these diseases, without major modifications of the self-renewal and proliferation capacities for JAK2V617F SCID repopulating cells. These experiments provide new insights into the pathogenesis of JAK2V617F MPD and demonstrate that a JAK2 inhibitor needs to target the HSC compartment for optimal disease control in classical MPD.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.