Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jyoti Nangalia is active.

Publication


Featured researches published by Jyoti Nangalia.


The New England Journal of Medicine | 2013

Somatic CALR Mutations in Myeloproliferative Neoplasms with Nonmutated JAK2

Jyoti Nangalia; Gunes Gundem; Edward Avezov; Jingjin Li; Karoline Kollmann; Athar Aziz; Jonathan Hinton; Inigo Martincorena; P. Van Loo; Paola Guglielmelli; Patrick Tarpey; Keiran Raine; Stuart McLaren; M. Bianchi; Yvonne Silber; D. Dimitropoulou; David Bloxham; Laura Mudie; Mark Maddison; Ben Robinson; Clodagh Keohane; Cathy MacLean; Katherine L. Hill; Kim H. Orchard; Sudhir Tauro; Mel Greaves; David G. Bowen; David Ron; Elli Papaemmanuil

BACKGROUND Somatic mutations in the Janus kinase 2 gene (JAK2) occur in many myeloproliferative neoplasms, but the molecular pathogenesis of myeloproliferative neoplasms with nonmutated JAK2 is obscure, and the diagnosis of these neoplasms remains a challenge. METHODS We performed exome sequencing of samples obtained from 151 patients with myeloproliferative neoplasms. The mutation status of the gene encoding calreticulin (CALR) was assessed in an additional 1345 hematologic cancers, 1517 other cancers, and 550 controls. We established phylogenetic trees using hematopoietic colonies. We assessed calreticulin subcellular localization using immunofluorescence and flow cytometry. RESULTS Exome sequencing identified 1498 mutations in 151 patients, with medians of 6.5, 6.5, and 13.0 mutations per patient in samples of polycythemia vera, essential thrombocythemia, and myelofibrosis, respectively. Somatic CALR mutations were found in 70 to 84% of samples of myeloproliferative neoplasms with nonmutated JAK2, in 8% of myelodysplasia samples, in occasional samples of other myeloid cancers, and in none of the other cancers. A total of 148 CALR mutations were identified with 19 distinct variants. Mutations were located in exon 9 and generated a +1 base-pair frameshift, which would result in a mutant protein with a novel C-terminal. Mutant calreticulin was observed in the endoplasmic reticulum without increased cell-surface or Golgi accumulation. Patients with myeloproliferative neoplasms carrying CALR mutations presented with higher platelet counts and lower hemoglobin levels than patients with mutated JAK2. Mutation of CALR was detected in hematopoietic stem and progenitor cells. Clonal analyses showed CALR mutations in the earliest phylogenetic node, a finding consistent with its role as an initiating mutation in some patients. CONCLUSIONS Somatic mutations in the endoplasmic reticulum chaperone CALR were found in a majority of patients with myeloproliferative neoplasms with nonmutated JAK2. (Funded by the Kay Kendall Leukaemia Fund and others.).


eLife | 2014

Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer.

Young Seok Ju; Ludmil B. Alexandrov; Moritz Gerstung; Inigo Martincorena; Serena Nik-Zainal; Manasa Ramakrishna; Helen Davies; Elli Papaemmanuil; Gunes Gundem; Adam Shlien; Niccolo Bolli; Sam Behjati; Patrick Tarpey; Jyoti Nangalia; C E Massie; Adam Butler; J Teague; George S. Vassiliou; Anthony R. Green; M Q Du; Ashwin Unnikrishnan; John E. Pimanda; Bin Tean Teh; Nikhil C. Munshi; Mel Greaves; Paresh Vyas; Adel K. El-Naggar; Thomas Santarius; V P Collins; Richard Grundy

Recent sequencing studies have extensively explored the somatic alterations present in the nuclear genomes of cancers. Although mitochondria control energy metabolism and apoptosis, the origins and impact of cancer-associated mutations in mtDNA are unclear. In this study, we analyzed somatic alterations in mtDNA from 1675 tumors. We identified 1907 somatic substitutions, which exhibited dramatic replicative strand bias, predominantly C > T and A > G on the mitochondrial heavy strand. This strand-asymmetric signature differs from those found in nuclear cancer genomes but matches the inferred germline process shaping primate mtDNA sequence content. A number of mtDNA mutations showed considerable heterogeneity across tumor types. Missense mutations were selectively neutral and often gradually drifted towards homoplasmy over time. In contrast, mutations resulting in protein truncation undergo negative selection and were almost exclusively heteroplasmic. Our findings indicate that the endogenous mutational mechanism has far greater impact than any other external mutagens in mitochondria and is fundamentally linked to mtDNA replication. DOI: http://dx.doi.org/10.7554/eLife.02935.001


Nature Communications | 2015

Genetic variation at MECOM , TERT , JAK2 and HBS1L-MYB predisposes to myeloproliferative neoplasms

William Tapper; Amy V. Jones; Robert Kralovics; Ashot S. Harutyunyan; Katerina Zoi; William Leung; Anna L. Godfrey; Paola Guglielmelli; Alison Callaway; Daniel Ward; Paula Aranaz; Helen E. White; Katherine Waghorn; Feng Lin; Andrew Chase; E. Joanna Baxter; Cathy MacLean; Jyoti Nangalia; Edwin Chen; Paul Evans; Michael Short; Andrew Jack; Louise Wallis; David Oscier; Andrew S Duncombe; Anna Schuh; Adam Mead; Michael Griffiths; Joanne Ewing; Rosemary E. Gale

Clonal proliferation in myeloproliferative neoplasms (MPN) is driven by somatic mutations in JAK2, CALR or MPL, but the contribution of inherited factors is poorly characterized. Using a three-stage genome-wide association study of 3,437 MPN cases and 10,083 controls, we identify two SNPs with genome-wide significance in JAK2V617F-negative MPN: rs12339666 (JAK2; meta-analysis P=1.27 × 10−10) and rs2201862 (MECOM; meta-analysis P=1.96 × 10−9). Two additional SNPs, rs2736100 (TERT) and rs9376092 (HBS1L/MYB), achieve genome-wide significance when including JAK2V617F-positive cases. rs9376092 has a stronger effect in JAK2V617F-negative cases with CALR and/or MPL mutations (Breslow–Day P=4.5 × 10−7), whereas in JAK2V617F-positive cases rs9376092 associates with essential thrombocythemia (ET) rather than polycythemia vera (allelic χ2 P=7.3 × 10−7). Reduced MYB expression, previously linked to development of an ET-like disease in model systems, associates with rs9376092 in normal myeloid cells. These findings demonstrate that multiple germline variants predispose to MPN and link constitutional differences in MYB expression to disease phenotype.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Molecular basis of thrombin recognition by protein C inhibitor revealed by the 1.6-Å structure of the heparin-bridged complex

Wei Li; Ty E. Adams; Jyoti Nangalia; Charles T. Esmon; James A. Huntington

Protein C inhibitor (PCI) is a serpin with many roles in biology, including a dual role as pro- and anticoagulant in blood. The protease specificity and local function of PCI depend on its interaction with cofactors such as heparin-like glycosaminoglycans (GAGs) and thrombomodulin (TM). Both cofactors significantly increase the rate of thrombin inhibition, but GAGs serve to promote the anticoagulant activity of PCI, and TM promotes its procoagulant function. To gain insight into how PCI recognition of thrombin is aided by these cofactors, we determined a crystallographic structure of the Michaelis complex of PCI, thrombin, and heparin to 1.6 Å resolution. Thrombin interacts with PCI in an unusual fashion that depends on the length of PCIs reactive center loop (RCL) to align the heparin-binding sites of the two proteins. The principal exosite contact is engendered by movement of thrombins 60-loop in response to the unique P2 Phe of PCI. This mechanism of communication between the active site of thrombin and its recognition exosite is previously uncharacterized and may relate to other thrombin substrate–cofactor interactions. The cofactor activity of heparin thus depends on the formation of a heparin-bridged Michaelis complex and substrate-induced exosite contacts. We also investigated the cofactor effect of TM, establishing that TM bridges PCI to thrombin through additional direct interactions. A model of the PCI–thrombin–TM complex was built and evaluated by mutagenesis and suggests distinct binding sites for heparin and TM on PCI. These data significantly improve our understanding of the cofactor-dependent roles of PCI in hemostasis.


Leukemia | 2014

BET protein inhibition shows efficacy against JAK2V617F driven neoplasms

Bs Wyspiańska; Andrew J. Bannister; Isaia Barbieri; Jyoti Nangalia; Anna L. Godfrey; Fernando J. Calero-Nieto; Samuel Robson; I Rioja; Juan Li; M Wiese; Ester Cannizzaro; Mark A. Dawson; Brian J. P. Huntly; Rab K. Prinjha; Anthony R. Green; Berthold Göttgens; Tony Kouzarides

Small molecule inhibition of the BET family of proteins, which bind acetylated lysines within histones, has been shown to have a marked therapeutic benefit in pre-clinical models of mixed lineage leukemia (MLL) fusion protein-driven leukemias. Here, we report that I-BET151, a highly specific BET family bromodomain inhibitor, leads to growth inhibition in a human erythroleukemic (HEL) cell line as well as in erythroid precursors isolated from polycythemia vera patients. One of the genes most highly downregulated by I-BET151 was LMO2, an important oncogenic regulator of hematopoietic stem cell development and erythropoiesis. We previously reported that LMO2 transcription is dependent upon Janus kinase 2 (JAK2) kinase activity in HEL cells. Here, we show that the transcriptional changes induced by a JAK2 inhibitor (TG101209) and I-BET151 in HEL cells are significantly over-lapping, suggesting a common pathway of action. We generated JAK2 inhibitor resistant HEL cells and showed that these retain sensitivity to I-BET151. These data highlight I-BET151 as a potential alternative treatment against myeloproliferative neoplasms driven by constitutively active JAK2 kinase.


Nature Genetics | 2014

Inactivating CUX1 mutations promote tumorigenesis

Chi C. Wong; Inigo Martincorena; Alistair G. Rust; Mamunur Rashid; Constantine Alifrangis; Ludmil B. Alexandrov; Jessamy Tiffen; Christina Kober; Anthony R. Green; Charlie E. Massie; Jyoti Nangalia; Stella Lempidaki; Hartmut Döhner; Konstanze Döhner; Sarah Bray; Ultan McDermott; Elli Papaemmanuil; Peter J. Campbell; David J. Adams

A major challenge in cancer genetics is to determine which low-frequency somatic mutations are drivers of tumorigenesis. Here we interrogate the genomes of 7,651 diverse human cancers and find inactivating mutations in the homeodomain transcription factor gene CUX1 (cut-like homeobox 1) in ∼1–5% of various tumors. Meta-analysis of CUX1 mutational status in 2,519 cases of myeloid malignancies reveals disruptive mutations associated with poor survival, highlighting the clinical significance of CUX1 loss. In parallel, we validate CUX1 as a bona fide tumor suppressor using mouse transposon-mediated insertional mutagenesis and Drosophila cancer models. We demonstrate that CUX1 deficiency activates phosphoinositide 3-kinase (PI3K) signaling through direct transcriptional downregulation of the PI3K inhibitor PIK3IP1 (phosphoinositide-3-kinase interacting protein 1), leading to increased tumor growth and susceptibility to PI3K-AKT inhibition. Thus, our complementary approaches identify CUX1 as a pan-driver of tumorigenesis and uncover a potential strategy for treating CUX1-mutant tumors.


The Lancet Haematology | 2017

Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial

Ruben A. Mesa; Alessandro M. Vannucchi; Adam Mead; Miklós Egyed; Anita Szoke; Aleksandr Suvorov; János Jakucs; Andrew C. Perkins; Ritam Prasad; Jiri Mayer; Judit Demeter; Peter Ganly; Jack W. Singer; Huafeng Zhou; James P. Dean; Peter te Boekhorst; Jyoti Nangalia; Jean-Jacques Kiladjian; Claire N. Harrison

BACKGROUND Available therapies for myelofibrosis can exacerbate cytopenias and are not indicated for patients with severe thrombocytopenia. Pacritinib, which inhibits both JAK2 and FLT3, induced spleen responses with limited myelosuppression in phase 1/2 trials. We aimed to assess the efficacy and safety of pacritinib versus best available therapy in patients with myelofibrosis irrespective of baseline cytopenias. METHODS This international, multicentre, randomised, phase 3 trial (PERSIST-1) was done at 67 sites in 12 countries. Patients with higher-risk myelofibrosis (with no exclusions for baseline anaemia or thrombocytopenia) were randomly assigned (2:1) to receive oral pacritinib 400 mg once daily or best available therapy (BAT) excluding JAK2 inhibitors until disease progression or unacceptable toxicity. Randomisation was stratified by risk category, platelet count, and region. Treatment assignments were known to investigators, site personnel, patients, clinical monitors, and pharmacovigilance personnel. The primary endpoint was spleen volume reduction (SVR) of 35% or more from baseline to week 24 in the intention-to-treat population as assessed by blinded, centrally reviewed MRI or CT. We did safety analyses in all randomised patients who received either treatment. Here we present the final data. This trial is registered with ClinicalTrials.gov, number NCT01773187. FINDINGS Between Jan 8, 2013, and Aug 1, 2014, 327 patients were randomly assigned to pacritinib (n=220) or BAT (n=107). Median follow-up was 23·2 months (IQR 14·8-28·7). At week 24, the primary endpoint of SVR of 35% or more was achieved by 42 (19%) patients in the pacritinib group versus five (5%) patients in the BAT group (p=0·0003). 90 patients in the BAT group crossed over to receive pacritinib at a median of 6·3 months (IQR 5·8-6·7). The most common grade 3-4 adverse events through week 24 were anaemia (n=37 [17%]), thrombocytopenia (n=26 [12%]), and diarrhoea (n=11 [5%]) in the pacritinib group, and anaemia (n=16 [15%]), thrombocytopenia (n=12 [11%]), dyspnoea (n=3 [3%]), and hypotension (n=3 [3%]) in the BAT group. The most common serious adverse events that occurred through week 24 were anaemia (10 [5%]), cardiac failure (5 [2%]), pyrexia (4 [2%]), and pneumonia (4 [2%]) with pacritinib, and anaemia (5 [5%]), sepsis (2 [2%]), and dyspnoea (2 [2%]) with BAT. Deaths due to adverse events were observed in 27 (12%) patients in the pacritinib group and 14 (13%) patients in the BAT group throughout the duration of the study. INTERPRETATION Pacritinib therapy was well tolerated and induced significant and sustained SVR and symptom reduction, even in patients with severe baseline cytopenias. Pacritinib could be a treatment option for patients with myelofibrosis, including those with baseline cytopenias for whom options are particularly limited. FUNDING CTI BioPharma Corp.


Leukemia Research | 2015

Evaluation of methods to detect CALR mutations in myeloproliferative neoplasms.

Amy V. Jones; Daniel Ward; Matthew Lyon; William Leung; Alison Callaway; Andrew Chase; Carolyn L. Dent; Helen E. White; Hans G. Drexler; Jyoti Nangalia; Chris Mattocks; Nicholas C.P. Cross

The recent discovery of somatically acquired CALR mutations in a substantial proportion of patients with myeloproliferative neoplasms has provided a new marker of clonal disease, advancing both diagnosis and prognosis in these previously difficult to characterise disorders. The mutations, which can be challenging to detect on a routine basis, are heterogeneous insertions/deletions (indels) in exon 9 with mutant allele burden that vary substantially between patients. We evaluated four genetic screening methods for their ability to detect a series of different CALR mutations; Sanger sequencing, fragment analysis PCR, high resolution melt (HRM) and targeted next generation sequencing (NGS). The limit of detection (LoD) of each assay was tested using serial dilution series made with DNA from CALR positive sample DNA and a cell line, MARIMO, found to carry a heterozygous 61 nucleotide CALR deletion. All methods were capable of detecting each mutation; HRM and fragment analysis PCR were better at detecting low mutation levels compared to Sanger sequencing but targeted NGS had the lowest LoD at a 1% mutation burden.


Blood | 2014

JAK2V617F homozygosity drives a phenotypic switch in myeloproliferative neoplasms, but is insufficient to sustain disease

Juan Li; David G. Kent; Anna L. Godfrey; Harriet Manning; Jyoti Nangalia; Athar Aziz; Edwin Chen; Kourosh Saeb-Parsy; Juergen Fink; Rachel Sneade; Tina L. Hamilton; Dean C. Pask; Yvonne Silber; Xiaodong Zhao; Cedric Ghevaert; Pentao Liu; Anthony R. Green

Genomic regions of acquired uniparental disomy (UPD) are common in malignancy and frequently harbor mutated oncogenes. Homozygosity for such gain-of-function mutations is thought to modulate tumor phenotype, but direct evidence has been elusive. Polycythemia vera (PV) and essential thrombocythemia (ET), 2 subtypes of myeloproliferative neoplasms, are associated with an identical acquired JAK2V617F mutation but the mechanisms responsible for distinct clinical phenotypes remain unclear. We provide direct genetic evidence and demonstrate that homozygosity for human JAK2V617F in knock-in mice results in a striking phenotypic switch from an ET-like to PV-like phenotype. The resultant erythrocytosis is driven by increased numbers of early erythroid progenitors and enhanced erythroblast proliferation, whereas reduced platelet numbers are associated with impaired platelet survival. JAK2V617F-homozygous mice developed a severe hematopoietic stem cell defect, suggesting that additional lesions are needed to sustain clonal expansion. Together, our results indicate that UPD for 9p plays a causal role in the PV phenotype in patients as a consequence of JAK2V617F homozygosity. The generation of a JAK2V617F allelic series of mice with a dose-dependent effect on hematopoiesis provides a powerful model for studying the consequences of mutant JAK2 homozygosity.


Genome Research | 2015

Frequent somatic transfer of mitochondrial DNA into the nuclear genome of human cancer cells

Young Seok Ju; Jose M. C. Tubio; William Mifsud; Beiyuan Fu; Helen Davies; Manasa Ramakrishna; Yilong Li; Lucy R. Yates; Gunes Gundem; Patrick Tarpey; Sam Behjati; Elli Papaemmanuil; Sancha Martin; Anthony Fullam; Moritz Gerstung; Jyoti Nangalia; Anthony R. Green; Carlos Caldas; Åke Borg; Andrew Tutt; Ming Ta Michael Lee; Laura J. van't Veer; Benita K T Tan; Samuel Aparicio; Paul N. Span; John W.M. Martens; Stian Knappskog; Anne Vincent-Salomon; Anne Lise Børresen-Dale; Jórunn Erla Eyfjörd

Mitochondrial genomes are separated from the nuclear genome for most of the cell cycle by the nuclear double membrane, intervening cytoplasm, and the mitochondrial double membrane. Despite these physical barriers, we show that somatically acquired mitochondrial-nuclear genome fusion sequences are present in cancer cells. Most occur in conjunction with intranuclear genomic rearrangements, and the features of the fusion fragments indicate that nonhomologous end joining and/or replication-dependent DNA double-strand break repair are the dominant mechanisms involved. Remarkably, mitochondrial-nuclear genome fusions occur at a similar rate per base pair of DNA as interchromosomal nuclear rearrangements, indicating the presence of a high frequency of contact between mitochondrial and nuclear DNA in some somatic cells. Transmission of mitochondrial DNA to the nuclear genome occurs in neoplastically transformed cells, but we do not exclude the possibility that some mitochondrial-nuclear DNA fusions observed in cancer occurred years earlier in normal somatic cells.

Collaboration


Dive into the Jyoti Nangalia's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna L. Godfrey

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Claire N. Harrison

Guy's and St Thomas' NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Elli Papaemmanuil

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter J. Campbell

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacob Grinfeld

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge