Network


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

Hotspot


Dive into the research topics where Nisha Limaye is active.

Publication


Featured researches published by Nisha Limaye.


Nature Genetics | 2011

Somatic mosaic IDH1 and IDH2 mutations are associated with enchondroma and spindle cell hemangioma in Ollier disease and Maffucci syndrome

Twinkal C. Pansuriya; Ronald van Eijk; Pio D'Adamo; Maayke A.J.H. van Ruler; Marieke L. Kuijjer; Jan Oosting; Anne-Marie Cleton-Jansen; Jolieke G. van Oosterwijk; Sofie L. J. Verbeke; Danielle Meijer; Tom van Wezel; Karolin Hansén Nord; Luca Sangiorgi; Berkin Toker; Bernadette Liegl-Atzwanger; Mikel San-Julian; Raf Sciot; Nisha Limaye; Lars-Gunnar Kindblom; Soeren Daugaard; Catherine Godfraind; Laurence M. Boon; Miikka Vikkula; Kyle C. Kurek; Karoly Szuhai; Pim J. French; Judith V. M. G. Bovée

Ollier disease and Maffucci syndrome are non-hereditary skeletal disorders characterized by multiple enchondromas (Ollier disease) combined with spindle cell hemangiomas (Maffucci syndrome). We report somatic heterozygous mutations in IDH1 (c.394C>T encoding an R132C substitution and c.395G>A encoding an R132H substitution) or IDH2 (c.516G>C encoding R172S) in 87% of enchondromas (benign cartilage tumors) and in 70% of spindle cell hemangiomas (benign vascular lesions). In total, 35 of 43 (81%) subjects with Ollier disease and 10 of 13 (77%) with Maffucci syndrome carried IDH1 (98%) or IDH2 (2%) mutations in their tumors. Fourteen of 16 subjects had identical mutations in separate lesions. Immunohistochemistry to detect mutant IDH1 R132H protein suggested intraneoplastic and somatic mosaicism. IDH1 mutations in cartilage tumors were associated with hypermethylation and downregulated expression of several genes. Mutations were also found in 40% of solitary central cartilaginous tumors and in four chondrosarcoma cell lines, which will enable functional studies to assess the role of IDH1 and IDH2 mutations in tumor formation.


Nature Medicine | 2008

Suppressed NFAT-dependent VEGFR1 expression and constitutive VEGFR2 signaling in infantile hemangioma

Masatoshi Jinnin; Damian Medici; Lucy Park; Nisha Limaye; Yanqiu Liu; Elisa Boscolo; Joyce Bischoff; Miikka Vikkula; Eileen Boye; Björn Olsen

Infantile hemangiomas are localized and rapidly growing regions of disorganized angiogenesis. We show that expression of vascular endothelial growth factor receptor-1 (VEGFR1) in hemangioma endothelial cells (hemECs) and hemangioma tissue is markedly reduced compared to controls. Low VEGFR1 expression in hemECs results in VEGF-dependent activation of VEGFR2 and downstream signaling pathways. In hemECs, transcription of the gene encoding VEGFR1 (FLT1) is dependent on nuclear factor of activated T cells (NFAT). Low VEGFR1 expression in hemECs is caused by reduced activity of a pathway involving β1 integrin, the integrin-like receptor tumor endothelial marker-8 (TEM8), VEGFR2 and NFAT. In a subset of individuals with hemangioma, we found missense mutations in the genes encoding VEGFR2 (KDR) and TEM8 (ANTXR1). These mutations result in increased interactions among VEGFR2, TEM8 and β1 integrin proteins and in inhibition of integrin activity. Normalization of the constitutive VEGFR2 signaling in hemECs with soluble VEGFR1 or antibodies that neutralize VEGF or stimulate β1 integrin suggests that local administration of these or similar agents may be effective in hemangioma treatment.


European Journal of Human Genetics | 2010

Hereditary cutaneomucosal venous malformations are caused by TIE2 mutations with widely variable hyper-phosphorylating effects

Vinciane Wouters; Nisha Limaye; Mélanie Uebelhoer; Alexandre Irrthum; Laurence M. Boon; John B. Mulliken; Odile Enjolras; Eulalia Baselga; Jonathan Berg; Anne Dompmartin; Sten A Ivarsson; Loshan Kangesu; Yves Lacassie; Jill Murphy; Ahmad S. Teebi; Anthony J. Penington; Paul N. M. A. Rieu; Miikka Vikkula

Mutations in the angiopoietin receptor TIE2/TEK have been identified as the cause for autosomal dominantly inherited cutaneomucosal venous malformation (VMCM). Thus far, two specific germline substitutions (R849W and Y897S), located in the kinase domain of TIE2, have been reported in five families. The mutations result in a fourfold increase in ligand-independent phosphorylation of the receptor. Here, we report 12 new families with TEK mutations. Although the phenotype is primarily characterized by small multifocal cutaneous vascular malformations, many affected members also have mucosal lesions. In addition, cardiac malformations are observed in some families. Six of the identified mutations are new, with three located in the tyrosine kinase domain, two in the kinase insert domain, and another in the carboxy terminal tail. The remaining six are R849W substitutions. Overexpression of the new mutants resulted in ligand-independent hyperphosphorylation of the receptor, suggesting this is a general feature of VMCM-causative TIE2 mutations. Moreover, variation in the level of activation demonstrates, to the best of our knowledge for the first time, that widely differing levels of chronic TIE2 hyperphosphorylation are tolerated in the heterozygous state, and are compatible with normal endothelial cell function except in the context of highly localized areas of lesion pathogenesis.


Human Molecular Genetics | 2009

From germline towards somatic mutations in the pathophysiology of vascular anomalies

Nisha Limaye; Laurence M. Boon; Miikka Vikkula

The localized structural abnormalities that arise during vasculogenesis, angiogenesis and lymphangiogenesis, the developmental processes which give rise to the adult vasculature, are collectively termed vascular anomalies. The last 2 years have seen an explosion of studies that underscore paradominant inheritance, the combination of inherited changes with somatic second-hits to the same genes, as underlying rare familial forms. Moreover, local, somatic genetic defects that cause some of the common sporadic forms of these malformations have been unraveled. This highlights the importance of assessing for tissue-based genetic changes, especially acquired genetic changes, as possible pathophysiological causes, which have been largely overlooked except in the area of cancer research. Large-scale somatic screens will therefore be essential in uncovering the nature and prevalence of such changes, and their downstream effects. The identification of disease genes combined with exhaustive, precise clinical delineations of the entire spectra of associated phenotypes guides better management and genetic counseling. Such a synthesis of information on functional and phenotypic effects will enable us to make and use animal models to test less invasive, targeted, perhaps locally administered, biological therapies.


American Journal of Human Genetics | 2015

Somatic Activating PIK3CA Mutations Cause Venous Malformation

Nisha Limaye; Jaakko Kangas; Antonella Mendola; Catherine Godfraind; Matthieu J. Schlögel; Raphaël Helaers; Lauri Eklund; Laurence M. Boon; Miikka Vikkula

Somatic mutations in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2, cause more than half of sporadically occurring unifocal venous malformations (VMs). Here, we report that somatic mutations in PIK3CA, the gene encoding the catalytic p110α subunit of PI3K, cause 54% (27 out of 50) of VMs with no detected TEK mutation. The hotspot mutations c.1624G>A, c.1633G>A, and c.3140A>G (p.Glu542Lys, p.Glu545Lys, and p.His1047Arg), frequent in PIK3CA-associated cancers, overgrowth syndromes, and lymphatic malformation (LM), account for >92% of individuals who carry mutations. Like VM-causative mutations in TEK, the PIK3CA mutations cause chronic activation of AKT, dysregulation of certain important angiogenic factors, and abnormal endothelial cell morphology when expressed in human umbilical vein endothelial cells (HUVECs). The p110α-specific inhibitor BYL719 restores all abnormal phenotypes tested, in PIK3CA- as well as TEK-mutant HUVECs, demonstrating that they operate via the same pathogenic pathways. Nevertheless, significant genotype-phenotype correlations in lesion localization and histology are observed between individuals with mutations in PIK3CA versus TEK, pointing to gene-specific effects.


Journal of Clinical Investigation | 2015

Rapamycin improves TIE2-mutated venous malformation in murine model and human subjects.

Elisa Boscolo; Nisha Limaye; Lan Huang; Kyu-Tae Kang; Julie Soblet; Mélanie Uebelhoer; Antonella Mendola; Marjut Nätynki; Emmanuel Seront; Sophie Dupont; Jennifer Hammer; Catherine Legrand; Carlo Brugnara; Lauri Eklund; Miikka Vikkula; Joyce Bischoff; Laurence M. Boon

Venous malformations (VMs) are composed of ectatic veins with scarce smooth muscle cell coverage. Activating mutations in the endothelial cell tyrosine kinase receptor TIE2 are a common cause of these lesions. VMs cause deformity, pain, and local intravascular coagulopathy, and they expand with time. Targeted pharmacological therapies are not available for this condition. Here, we generated a model of VMs by injecting HUVECs expressing the most frequent VM-causing TIE2 mutation, TIE2-L914F, into immune-deficient mice. TIE2-L914F-expressing HUVECs formed VMs with ectatic blood-filled channels that enlarged over time. We tested both rapamycin and a TIE2 tyrosine kinase inhibitor (TIE2-TKI) for their effects on murine VM expansion and for their ability to inhibit mutant TIE2 signaling. Rapamycin prevented VM growth, while TIE2-TKI had no effect. In cultured TIE2-L914F-expressing HUVECs, rapamycin effectively reduced mutant TIE2-induced AKT signaling and, though TIE2-TKI did target the WT receptor, it only weakly suppressed mutant-induced AKT signaling. In a prospective clinical pilot study, we analyzed the effects of rapamycin in 6 patients with difficult-to-treat venous anomalies. Rapamycin reduced pain, bleeding, lesion size, functional and esthetic impairment, and intravascular coagulopathy. This study provides a VM model that allows evaluation of potential therapeutic strategies and demonstrates that rapamycin provides clinical improvement in patients with venous malformation.


Molecular Syndromology | 2013

Variable Somatic TIE2 Mutations in Half of Sporadic Venous Malformations.

Julie Soblet; Nisha Limaye; Mélanie Uebelhoer; Laurence M. Boon; Miikka Vikkula

Venous malformations (VMs) are the most frequent vascular malformations referred to specialized vascular anomaly centers. A rare (1-2%) familial form, termed cutaneomucosal venous malformation (VMCM), is caused by gain-of-function mutations in TIE2. More recently, sporadic VMs, characterized by the presence of large unifocal lesions, were shown to be caused by somatic mutations in TIE2. These include a frequent L914F change, and a series of double mutations in cis. All of which cause ligand-independent receptor hyperphosphorylation in vitro. Here, we expanded our study to assess the range of mutations that cause sporadic VM. To test for somatic changes, we screened the entire coding region of TIE2 in cDNA from resected VMs by direct sequencing. We detected TIE2 mutations in 17/30 (56.7%) of the samples. In addition to previously detected mutations, we identified 7 novel somatic intracellular TIE2 mutations in sporadic VMs, including 3 that cause premature protein truncation.


Human Molecular Genetics | 2013

Venous malformation-causative TIE2 mutations mediate an AKT-dependent decrease in PDGFB

Mélanie Uebelhoer; Marjut Nätynki; Jaakko Kangas; Antonella Mendola; Ha-Long Nguyen; Julie Soblet; Catherine Godfraind; Laurence M. Boon; Lauri Eklund; Nisha Limaye; Miikka Vikkula

Mutations in the endothelial cell (EC) tyrosine kinase receptor TIE2 cause inherited and sporadic forms of venous malformation. The recurrent somatic mutation L914F and common germline mutation R849W differ in terms of phosphorylation level, as well as sub-cellular localization and trafficking of the receptor. Previous studies have shed light on certain pathogenic properties of R849W, but the mechanisms of action of L914F are unknown. We used global gene expression profiling to study the effects of L914F on ECs. We found that L914F strongly dysregulates genes involved in vascular development, cell migration and extracellular matrix processing, while R849W has weak effects. We also demonstrate, for the first time, that TIE2-mutant ECs are deficient in the production of PDGFB, both in vitro and ex vivo in patient tissues. This defect is mediated by the chronic, ligand-independent activation of AKT by the mutant receptors. Inadequate secretion of the major mural cell attractant likely plays an important role in the development of abnormal vascular channels, contributing to the characteristic paucity of surrounding vascular smooth muscle cells.


Journal of Investigative Dermatology | 2017

Blue Rubber Bleb Nevus (BRBN) Syndrome Is Caused by Somatic TEK (TIE2) Mutations.

Julie Soblet; Jaakko Kangas; Marjut Nätynki; Antonella Mendola; Raphaël Helaers; Mélanie Uebelhoer; Mika Kaakinen; Maria R. Cordisco; Anne Dompmartin; Odile Enjolras; Simon Holden; Alan D. Irvine; Loshan Kangesu; Christine Léauté-Labrèze; Agustina Lanoel; Zerina Lokmic; Saskia M. Maas; Maeve A. McAleer; Anthony J. Penington; Paul N. M. A. Rieu; Samira Syed; Carine van der Vleuten; Rosemarie Watson; Steven J. Fishman; John B. Mulliken; Lauri Eklund; Nisha Limaye; Laurence M. Boon; Miikka Vikkula

Blue rubber bleb nevus syndrome (Bean syndrome) is a rare, severe disorder of unknown cause, characterized by numerous cutaneous and internal venous malformations; gastrointestinal lesions are pathognomonic. We discovered somatic mutations in TEK, the gene encoding TIE2, in 15 of 17 individuals with blue rubber bleb nevus syndrome. Somatic mutations were also identified in five of six individuals with sporadically occurring multifocal venous malformations. In contrast to common unifocal venous malformation, which is most often caused by the somatic L914F TIE2 mutation, multifocal forms are predominantly caused by double (cis) mutations, that is, two somatic mutations on the same allele of the gene. Mutations are identical in all lesions from a given individual. T1105N-T1106P is recurrent in blue rubber bleb nevus, whereas Y897C-R915C is recurrent in sporadically occurring multifocal venous malformation: both cause ligand-independent activation of TIE2, and increase survival, invasion, and colony formation when expressed in human umbilical vein endothelial cells.


Human Molecular Genetics | 2015

Common and specific effects of TIE2 mutations causing venous malformations

Marjut Nätynki; Jaakko Kangas; Ilkka Miinalainen; Raija Sormunen; Riikka Pietilä; Julie Soblet; Laurence M. Boon; Miikka Vikkula; Nisha Limaye; Lauri Eklund

Venous malformations (VMs) are localized defects in vascular morphogenesis frequently caused by mutations in the gene for the endothelial tyrosine kinase receptor TIE2. Here, we report the analysis of a comprehensive collection of 22 TIE2 mutations identified in patients with VM, either as single amino acid substitutions or as double-mutations on the same allele. Using endothelial cell (EC) cultures, mouse models and ultrastructural analysis of tissue biopsies from patients, we demonstrate common as well as mutation-specific cellular and molecular features, on the basis of which mutations cluster into categories that correlate with data from genetic studies. Comparisons of double-mutants with their constituent single-mutant forms identified the pathogenic contributions of individual changes, and their compound effects. We find that defective receptor trafficking and subcellular localization of different TIE2 mutant forms occur via a variety of mechanisms, resulting in attenuated response to ligand. We also demonstrate, for the first time, that TIE2 mutations cause chronic activation of the MAPK pathway resulting in loss of normal EC monolayer due to extracellular matrix (ECM) fibronectin deficiency and leading to upregulation of plasminogen/plasmin proteolytic pathway. Corresponding EC and ECM irregularities are observed in affected tissues from mouse models and patients. Importantly, an imbalance between plasminogen activators versus inhibitors would also account for high d-dimer levels, a major feature of unknown cause that distinguishes VMs from other vascular anomalies.

Collaboration


Dive into the Nisha Limaye's collaboration.

Top Co-Authors

Avatar

Miikka Vikkula

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Laurence M. Boon

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

Mélanie Uebelhoer

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie Soblet

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Raphaël Helaers

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Vinciane Wouters

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonella Mendola

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge