Network


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

Hotspot


Dive into the research topics where Shoumo Bhattacharya is active.

Publication


Featured researches published by Shoumo Bhattacharya.


Cell | 2003

Cyclin E Ablation in the Mouse

Yan Geng; Qunyan Yu; Ewa Sicinska; Manjusri Das; Jürgen E. Schneider; Shoumo Bhattacharya; William Rideout; Roderick T. Bronson; Humphrey Gardner; Piotr Sicinski

E type cyclins (E1 and E2) are believed to drive cell entry into the S phase. It is widely assumed that the two E type cyclins are critically required for proliferation of all cell types. Here, we demonstrate that E type cyclins are largely dispensable for mouse development. However, endoreplication of trophoblast giant cells and megakaryocytes is severely impaired in the absence of cyclin E. Cyclin E-deficient cells proliferate actively under conditions of continuous cell cycling but are unable to reenter the cell cycle from the quiescent G(0) state. Molecular analyses revealed that cells lacking cyclin E fail to normally incorporate MCM proteins into DNA replication origins during G(0)-->S progression. We also found that cyclin E-deficient cells are relatively resistant to oncogenic transformation. These findings define a molecular function for E type cyclins in cell cycle reentry and reveal a differential requirement for cyclin E in normal versus oncogenic proliferation.


Cell | 2004

Mouse Development and Cell Proliferation in the Absence of D-Cyclins

Katarzyna Kozar; Maria A. Ciemerych; Vivienne I. Rebel; Hirokazu Shigematsu; Agnieszka Zagozdzon; Ewa Sicinska; Yan Geng; Qunyan Yu; Shoumo Bhattacharya; Roderick T. Bronson; Koichi Akashi; Piotr Sicinski

D-type cyclins (cyclins D1, D2, and D3) are regarded as essential links between cell environment and the core cell cycle machinery. We tested the requirement for D-cyclins in mouse development and in proliferation by generating mice lacking all D-cyclins. We found that these cyclin D1(-/-)D2(-/-)D3(-/-) mice develop until mid/late gestation and die due to heart abnormalities combined with a severe anemia. Our analyses revealed that the D-cyclins are critically required for the expansion of hematopoietic stem cells. In contrast, cyclin D-deficient fibroblasts proliferate nearly normally but show increased requirement for mitogenic stimulation in cell cycle re-entry. We found that the proliferation of cyclin D1(-/-)D2(-/-)D3(-/-) cells is resistant to the inhibition by p16(INK4a), but it critically depends on CDK2. Lastly, we found that cells lacking D-cyclins display reduced susceptibility to the oncogenic transformation. Our results reveal the presence of alternative mechanisms that allow cell cycle progression in a cyclin D-independent fashion.


Nature Genetics | 2001

Cardiac malformations, adrenal agenesis, neural crest defects and exencephaly in mice lacking Cited2, a new Tfap2 co-activator.

Simon D. Bamforth; José Bragança; Jyrki J. Eloranta; Jennifer N. Murdoch; Fatima I.R. Marques; Kamil R. Kranc; Hend Farza; Deborah J. Henderson; Helen C. Hurst; Shoumo Bhattacharya

The protein EP300 and its paralog CREBBP (CREB-binding protein) are ubiquitously expressed transcriptional co-activators and histone acetyl transferases. The gene EP300 is essential for normal cardiac and neural development, whereas CREBBP is essential for neurulation, hematopoietic differentiation, angiogenesis and skeletal and cardiac development. Mutations in CREBBP cause Rubinstein-Taybi syndrome, which is characterized by mental retardation, skeletal abnormalities and congenital cardiac defects. The CBP/p300-interacting transactivator with ED-rich tail 2 (CITED2) binds EP300 and CREBBP with high affinity and regulates gene transcription. Here we show that Cited2−/− embryos die with cardiac malformations, adrenal agenesis, abnormal cranial ganglia and exencephaly. The cardiac defects include atrial and ventricular septal defects, overriding aorta, double-outlet right ventricle, persistent truncus arteriosus and right-sided aortic arches. We find increased apoptosis in the midbrain region and a marked reduction in ErbB3-expressing neural crest cells in mid-embryogenesis. We show that CITED2 interacts with and co-activates all isoforms of transcription factor AP-2 (TFAP2). Transactivation by TFAP2 isoforms is defective in Cited2−/− embryonic fibroblasts and is rescued by ectopically expressed CITED2. As certain Tfap2 isoforms are essential in neural crest, neural tube and cardiac development, we propose that abnormal embryogenesis in mice lacking Cited2 results, at least in part, from its role as a Tfap2 co-activator.


Nature Genetics | 2004

Cited2 controls left-right patterning and heart development through a Nodal-Pitx2c pathway

Simon D. Bamforth; José Bragança; Cassandra R. Farthing; Jürgen E. Schneider; Carol Broadbent; Anna C. Michell; Kieran Clarke; Stefan Neubauer; Dominic P. Norris; Nigel A. Brown; Robert H. Anderson; Shoumo Bhattacharya

Malformations of the septum, outflow tract and aortic arch are the most common congenital cardiovascular defects and occur in mice lacking Cited2, a transcriptional coactivator of TFAP2. Here we show that Cited2−/− mice also develop laterality defects, including right isomerism, abnormal cardiac looping and hyposplenia, which are suppressed on a mixed genetic background. Cited2−/− mice lack expression of the Nodal target genes Pitx2c, Nodal and Ebaf in the left lateral plate mesoderm, where they are required for establishing laterality and cardiovascular development. CITED2 and TFAP2 were detected at the Pitx2c promoter in embryonic hearts, and they activate Pitx2c transcription in transient transfection assays. We propose that an abnormal Nodal-Pitx2c pathway represents a unifying mechanism for the cardiovascular malformations observed in Cited2−/− mice, and that such malformations may be the sole manifestation of a laterality defect.


Nature Genetics | 2011

Sox2 cooperates with Chd7 to regulate genes that are mutated in human syndromes

Erik Engelen; Umut Akinci; Jan Christian Bryne; Jun Hou; Cristina Gontan; Maaike Moen; Dorota Szumska; Christel Kockx; Wilfred van IJcken; Dick H. W. Dekkers; Jeroen Demmers; Erikjan Rijkers; Shoumo Bhattacharya; Sjaak Philipsen; Larysa Pevny; Frank Grosveld; Robbert J. Rottier; Boris Lenhard; Raymond A. Poot

The HMG-box transcription factor Sox2 plays a role throughout neurogenesis and also acts at other stages of development, as illustrated by the multiple organs affected in the anophthalmia syndrome caused by SOX2 mutations. Here we combined proteomic and genomic approaches to characterize gene regulation by Sox2 in neural stem cells. Chd7, a chromatin remodeling ATPase associated with CHARGE syndrome, was identified as a Sox2 transcriptional cofactor. Sox2 and Chd7 physically interact, have overlapping genome-wide binding sites and regulate a set of common target genes including Jag1, Gli3 and Mycn, genes mutated in Alagille, Pallister-Hall and Feingold syndromes, which show malformations also associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Regulation of disease-associated genes by a Sox2-Chd7 complex provides a plausible explanation for several malformations associated with SOX2 anophthalmia syndrome or CHARGE syndrome. Indeed, we found that Chd7-haploinsufficient embryos showed severely reduced expression of Jag1 in the developing inner ear.


American Journal of Human Genetics | 2012

Contribution of global rare copy-number variants to the risk of sporadic congenital heart disease.

Rachel Soemedi; Ian Wilson; Jamie Bentham; Rebecca Darlay; Ana Töpf; Diana Zelenika; Catherine Cosgrove; Kerry Setchfield; Chris Thornborough; Javier Granados-Riveron; Gillian M. Blue; Jeroen Breckpot; Stephen Hellens; Simon Zwolinkski; Elise Glen; Chrysovalanto Mamasoula; Thahira Rahman; Darroch Hall; Anita Rauch; Koenraad Devriendt; Marc Gewillig; John O’Sullivan; David S. Winlaw; Frances A. Bu’Lock; J. David Brook; Shoumo Bhattacharya; Mark Lathrop; Mauro Santibanez-Koref; Heather J. Cordell; Judith A. Goodship

Previous studies have shown that copy-number variants (CNVs) contribute to the risk of complex developmental phenotypes. However, the contribution of global CNV burden to the risk of sporadic congenital heart disease (CHD) remains incompletely defined. We generated genome-wide CNV data by using Illumina 660W-Quad SNP arrays in 2,256 individuals with CHD, 283 trio CHD-affected families, and 1,538 controls. We found association of rare genic deletions with CHD risk (odds ratio [OR] = 1.8, p = 0.0008). Rare deletions in study participants with CHD had higher gene content (p = 0.001) with higher haploinsufficiency scores (p = 0.03) than they did in controls, and they were enriched with Wnt-signaling genes (p = 1 × 10(-5)). Recurrent 15q11.2 deletions were associated with CHD risk (OR = 8.2, p = 0.02). Rare de novo CNVs were observed in ~5% of CHD trios; 10 out of 11 occurred on the paternally transmitted chromosome (p = 0.01). Some of the rare de novo CNVs spanned genes known to be involved in heart development (e.g., HAND2 and GJA5). Rare genic deletions contribute ~4% of the population-attributable risk of sporadic CHD. Second to previously described CNVs at 1q21.1, deletions at 15q11.2 and those implicating Wnt signaling are the most significant contributors to the risk of sporadic CHD. Rare de novo CNVs identified in CHD trios exhibit paternal origin bias.


BMC Developmental Biology | 2004

Identification of cardiac malformations in mice lacking Ptdsr using a novel high-throughput magnetic resonance imaging technique

Jürgen E. Schneider; Jens Böse; Simon D. Bamforth; Achim D. Gruber; Carol Broadbent; Kieran Clarke; Stefan Neubauer; Andreas Lengeling; Shoumo Bhattacharya

BackgroundCongenital heart defects are the leading non-infectious cause of death in children. Genetic studies in the mouse have been crucial to uncover new genes and signaling pathways associated with heart development and congenital heart disease. The identification of murine models of congenital cardiac malformations in high-throughput mutagenesis screens and in gene-targeted models is hindered by the opacity of the mouse embryo.ResultsWe developed and optimized a novel method for high-throughput multi-embryo magnetic resonance imaging (MRI). Using this approach we identified cardiac malformations in phosphatidylserine receptor (Ptdsr) deficient embryos. These included ventricular septal defects, double-outlet right ventricle, and hypoplasia of the pulmonary artery and thymus. These results indicate that Ptdsr plays a key role in cardiac development.ConclusionsOur novel multi-embryo MRI technique enables high-throughput identification of murine models for human congenital cardiopulmonary malformations at high spatial resolution. The technique can be easily adapted for mouse mutagenesis screens and, thus provides an important new tool for identifying new mouse models for human congenital heart diseases.


PLOS Genetics | 2010

A mutation in the mitochondrial fission gene Dnm1l leads to cardiomyopathy

Houman Ashrafian; Louise Docherty; Vincenzo C. Leo; Christopher Towlson; Monica Neilan; Violetta Steeples; Craig A. Lygate; Tertius Hough; Stuart Townsend; Debbie Williams; Sara Wells; Dominic P. Norris; Sarah Glyn-Jones; John M. Land; Ivana Barbaric; Zuzanne Lalanne; Paul Denny; Dorota Szumska; Shoumo Bhattacharya; Julian L. Griffin; Iain Hargreaves; Narcis Fernandez-Fuentes; Michael Cheeseman; Hugh Watkins; T. Neil Dear

Mutations in a number of genes have been linked to inherited dilated cardiomyopathy (DCM). However, such mutations account for only a small proportion of the clinical cases emphasising the need for alternative discovery approaches to uncovering novel pathogenic mutations in hitherto unidentified pathways. Accordingly, as part of a large-scale N-ethyl-N-nitrosourea mutagenesis screen, we identified a mouse mutant, Python, which develops DCM. We demonstrate that the Python phenotype is attributable to a dominant fully penetrant mutation in the dynamin-1-like (Dnm1l) gene, which has been shown to be critical for mitochondrial fission. The C452F mutation is in a highly conserved region of the M domain of Dnm1l that alters protein interactions in a yeast two-hybrid system, suggesting that the mutation might alter intramolecular interactions within the Dnm1l monomer. Heterozygous Python fibroblasts exhibit abnormal mitochondria and peroxisomes. Homozygosity for the mutation results in the death of embryos midway though gestation. Heterozygous Python hearts show reduced levels of mitochondria enzyme complexes and suffer from cardiac ATP depletion. The resulting energy deficiency may contribute to cardiomyopathy. This is the first demonstration that a defect in a gene involved in mitochondrial remodelling can result in cardiomyopathy, showing that the function of this gene is needed for the maintenance of normal cellular function in a relatively tissue-specific manner. This disease model attests to the importance of mitochondrial remodelling in the heart; similar defects might underlie human heart muscle disease.


Genes & Development | 2008

VACTERL/caudal regression/Currarino syndrome-like malformations in mice with mutation in the proprotein convertase Pcsk5

Dorota Szumska; Guido Pieles; Rachid Essalmani; M Bilski; Daniel Mesnard; K Kaur; Angela Franklyn; K El Omari; J Jefferis; Jamie Bentham; J M Taylor; Jürgen E. Schneider; Sebastian J. Arnold; P Johnson; Z Tymowska-Lalanne; David K. Stammers; Kieran Clarke; Stefan Neubauer; A Morris; Steve D.M. Brown; C Shaw-Smith; Armando Cama; Valeria Capra; J Ragoussis; Daniel B. Constam; Nabil G. Seidah; Annik Prat; Shoumo Bhattacharya

We have identified an ethylnitrosourea (ENU)-induced recessive mouse mutation (Vcc) with a pleiotropic phenotype that includes cardiac, tracheoesophageal, anorectal, anteroposterior patterning defects, exomphalos, hindlimb hypoplasia, a presacral mass, renal and palatal agenesis, and pulmonary hypoplasia. It results from a C470R mutation in the proprotein convertase PCSK5 (PC5/6). Compound mutants (Pcsk5(Vcc/null)) completely recapitulate the Pcsk5(Vcc/Vcc) phenotype, as does an epiblast-specific conditional deletion of Pcsk5. The C470R mutation ablates a disulfide bond in the P domain, and blocks export from the endoplasmic reticulum and proprotein convertase activity. We show that GDF11 is cleaved and activated by PCSK5A, but not by PCSK5A-C470R, and that Gdf11-deficient embryos, in addition to having anteroposterior patterning defects and renal and palatal agenesis, also have a presacral mass, anorectal malformation, and exomphalos. Pcsk5 mutation results in abnormal expression of several paralogous Hox genes (Hoxa, Hoxc, and Hoxd), and of Mnx1 (Hlxb9). These include known Gdf11 targets, and are necessary for caudal embryo development. We identified nonsynonymous mutations in PCSK5 in patients with VACTERL (vertebral, anorectal, cardiac, tracheoesophageal, renal, limb malformation OMIM 192350) and caudal regression syndrome, the phenotypic features of which resemble the mouse mutation. We propose that Pcsk5, at least in part via GDF11, coordinately regulates caudal Hox paralogs, to control anteroposterior patterning, nephrogenesis, skeletal, and anorectal development.


Cell Stem Cell | 2009

Cited2 Is an Essential Regulator of Adult Hematopoietic Stem Cells

Kamil R. Kranc; Hein Schepers; Neil P. Rodrigues; Simon D. Bamforth; Ellen Villadsen; Helen Ferry; Tiphaine Bouriez-Jones; Mikael Sigvardsson; Shoumo Bhattacharya; Sten Eirik W. Jacobsen; Tariq Enver

Summary The regulatory pathways necessary for the maintenance of adult hematopoietic stem cells (HSCs) remain poorly defined. By using loss-of-function approaches, we report a selective and cell-autonomous requirement for the p300/CBP-binding transcriptional coactivator Cited2 in adult HSC maintenance. Conditional deletion of Cited2 in the adult mouse results in loss of HSCs causing multilineage bone marrow failure and increased lethality. In contrast, conditional ablation of Cited2 after lineage specification in lymphoid and myeloid lineages has no impact on the maintenance of these lineages. Additional deletion of Ink4a/Arf (encoding p16Ink4a and p19Arf) or Trp53 (encoding p53, a downstream target of p19Arf) in a Cited2-deficient background restores HSC functionality and rescues mice from bone marrow failure. Furthermore, we show that the critical role of Cited2 in primitive hematopoietic cells is conserved in humans. Taken together, our studies provide genetic evidence that Cited2 selectively maintains adult HSC functions, at least in part, via Ink4a/Arf and Trp53.

Collaboration


Dive into the Shoumo Bhattacharya's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dorota Szumska

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

José Bragança

University of the Algarve

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chris Thornborough

University Hospitals of Leicester NHS Trust

View shared research outputs
Top Co-Authors

Avatar

Alan Hart

University of Edinburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge