Network


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

Hotspot


Dive into the research topics where Rikhia Chakraborty is active.

Publication


Featured researches published by Rikhia Chakraborty.


Blood | 2014

Mutually exclusive recurrent somatic mutations in MAP2K1 and BRAF support a central role for ERK activation in LCH pathogenesis.

Rikhia Chakraborty; Oliver A. Hampton; Xiaoyun Shen; Stephen J. Simko; Albert Shih; Harshal Abhyankar; Karen Phaik Har Lim; Kyle Covington; Lisa R. Trevino; Ninad Dewal; Donna M. Muzny; HarshaVardhan Doddapaneni; Jianhong Hu; Linghua Wang; Philip J. Lupo; M. John Hicks; Diana L. Bonilla; Karen C. Dwyer; Marie Luise Berres; Poulikos I. Poulikakos; Miriam Merad; Kenneth L. McClain; David A. Wheeler; Carl E. Allen; D. Williams Parsons

Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder characterized by lesions composed of pathological CD207(+) dendritic cells with an inflammatory infiltrate. BRAFV600E remains the only recurrent mutation reported in LCH. In order to evaluate the spectrum of somatic mutations in LCH, whole exome sequencing was performed on matched LCH and normal tissue samples obtained from 41 patients. Lesions from other histiocytic disorders, juvenile xanthogranuloma, Erdheim-Chester disease, and Rosai-Dorfman disease were also evaluated. All of the lesions from histiocytic disorders were characterized by an extremely low overall rate of somatic mutations. Notably, 33% (7/21) of LCH cases with wild-type BRAF and none (0/20) with BRAFV600E harbored somatic mutations in MAP2K1 (6 in-frame deletions and 1 missense mutation) that induced extracellular signal-regulated kinase (ERK) phosphorylation in vitro. Single cases of somatic mutations of the mitogen-activated protein kinase (MAPK) pathway genes ARAF and ERBB3 were also detected. The ability of MAPK pathway inhibitors to suppress MAPK kinase and ERK phosphorylation in cell culture and primary tumor models was dependent on the specific LCH mutation. The findings of this study support a model in which ERK activation is a universal end point in LCH arising from pathological activation of upstream signaling proteins.


Journal of Experimental Medicine | 2014

BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups

Marie-Luise Berres; Karen Phaik Har Lim; Tricia L. Peters; Jeremy Price; Hitoshi Takizawa; Hélène Salmon; Juliana Idoyaga; Albert Ruzo; Philip J. Lupo; M. John Hicks; Albert Shih; Stephen J. Simko; Harshal Abhyankar; Rikhia Chakraborty; Marylene Leboeuf; Monique F. Beltrao; Sergio A. Lira; Kenneth Matthew Heym; Björn E. Clausen; Venetia Bigley; Matthew Collin; Markus G. Manz; Kenneth L. McClain; Miriam Merad; Carl E. Allen

The Rockefeller University Press


Journal of Immunology | 2011

Activation of Wnt signaling arrests effector differentiation in human peripheral and cord blood-derived T lymphocytes

Sujatha Muralidharan; Patrick J. Hanley; Enli Liu; Rikhia Chakraborty; Catherine M. Bollard; Elizabeth J. Shpall; Cliona M. Rooney; Barbara Savoldo; John R. Rodgers; Gianpietro Dotti

30.00 J. Exp. Med. 2014 Vol. 211 No. 4 669-683 www.jem.org/cgi/doi/10.1084/jem.20130977 669 Langerhans cell histiocytosis (LCH) is characterized by inflammatory lesions that include pathological langerin+ DCs. LCH has pleotropic clinical presentations ranging from single lesions cured by curettage to potentially fatal multisystem disease. The first descriptions of LCH, including Hand-Schüller-Christian disease and Letter-Siwe disease, were based on anatomical location and extent of the lesions (Arceci, 1999). The diagnosis of high-risk LCH, defined by involvement of “risk organs” which include BM, liver, and spleen, conferred mortality rates >20%, where patients with disease limited to non-risk organs (low-risk LCH) had nearly 100% survival, CORRESPONDENCE Carl Allen: [email protected] OR Miriam Merad: [email protected]


The Journal of Pediatrics | 2014

Differentiating skin-limited and multisystem langerhans cell histiocytosis

Stephen J. Simko; Benjamin Garmezy; Harshal Abhyankar; Philip J. Lupo; Rikhia Chakraborty; Karen Phaik Har Lim; Albert Shih; M. John Hicks; Teresa S. Wright; Moise L. Levy; Kenneth L. McClain; Carl E. Allen

The canonical Wnt/β-catenin signaling pathway plays an important role in thymocyte development and T cell migration, but little is known about its role in naive-to-effector differentiation in human peripheral T cells. We show that activation of Wnt/β-catenin signaling arrests human peripheral blood and cord blood T lymphocytes in the naive stage and blocks their transition into functional T effector cells. Wnt signaling was induced in polyclonally activated human T cells by treatment either with the glycogen synthase kinase 3β inhibitor TWS119 or the physiological Wnt agonist Wnt-3a, and these T cells preserved a naive CD45RA+CD62L+ phenotype compared with control-activated T cells that progressed to a CD45RO+CD62L− effector phenotype, and this occurred in a TWS119 dose-dependent manner. TWS119-induced Wnt signaling reduced T cell expansion, as a result of a block in cell division, and impaired acquisition of T cell effector function, measured by degranulation and IFN-γ production in response to T cell activation. The block in T cell division may be attributed to the reduced IL-2Rα expression in TWS119-treated T cells that lowers their capacity to use autocrine IL-2 for expansion. Collectively, our data suggest that Wnt/β-catenin signaling is a negative regulator of naive-to-effector T cell differentiation in human T lymphocytes. The arrest in T cell differentiation induced by Wnt signaling might have relevant clinical applications such as to preserve the naive T cell compartment in Ag-specific T cells generated ex vivo for adoptive T cell immunotherapy.


Haematologica | 2013

Robust and cost effective expansion of human regulatory T cells highly functional in a xenograft model of graft versus host disease

Rikhia Chakraborty; Aruna Mahendravada; Serena K. Perna; Cliona M. Rooney; Helen E. Heslop; Juan F. Vera; Barbara Savoldo; Gianpietro Dotti

OBJECTIVE To identify features associated with multisystem involvement and therapeutic failure in patients with skin Langerhans cell histiocytosis (LCH). STUDY DESIGN We reviewed medical records of 71 consecutive patients with LCH with skin involvement evaluated at Texas Childrens Hospital and analyzed clinical features, laboratory results, and the presence of circulating cells with the BRAF-V600E mutation with respect to initial staging and clinical outcomes. RESULTS Skin disease in patients older than 18 months of age at diagnosis was associated with the presence of multisystem disease (OR, 9.65; 95% CI, 1.17-79.4). Forty percent of patients referred for presumed skin-limited LCH had underlying multisystem involvement, one-half of these with risk-organ involvement. Patients with skin-limited LCH had a 3-year progression-free survival of 89% after initial therapy, and none developed multisystem disease. Patients with skin/multisystem involvement had a 3-year progression-free survival of 44% with vinblastine/prednisone therapy, and risk-organ involvement did not correlate with failure to achieve nonactive disease. Circulating cells with BRAF-V600E were detected at higher frequency in patients with multisystem involvement (8 of 11 skin/multisystem vs 1 of 13 skin-limited; P = .002). CONCLUSION Skin-limited LCH necessitates infrequent therapeutic intervention and has a lower risk of progression relative to skin plus multisystem LCH. The less-aggressive clinical course and lack of circulating cells with the BRAF-V600E mutation in skin-limited LCH suggest a different mechanism of disease origin compared with multisystem or risk-organ disease.


Journal of Immunotherapy | 2012

Changes in Chemokine Receptor Expression of Regulatory T Cells after Ex Vivo Culture

Rikhia Chakraborty; Cliona M. Rooney; Gianpietro Dotti; Barbara Savoldo

The low frequency of naturally occurring regulatory T cells (nTregs) in peripheral blood and the suboptimal protocols available for their ex vivo expansion limit the development of clinical trials based on the adoptive transfer of these cells. We have, therefore, generated a simplified, robust and cost-effective platform for the large-scale expansion of nTregs using a gas permeable static culture flask (G-Rex) in compliance with Good Manufacturing Practice. More than 109 putative Tregs co-expressing CD25 and CD4 molecules (92±5%) and FoxP3 (69±19%) were obtained within 21 days of culture. Expanded Tregs showed potent regulatory activity in vitro (80±13% inhibition of CD8+ cell division) and in vivo (suppression or delay of graft-versus-host disease in a xenograft mouse model) indicating that the cost-effective and simplified production of nTregs we propose will facilitate the implementation of clinical trials based on their adoptive transfer.


Oncotarget | 2017

Activating MAPK1 (ERK2) mutation in an aggressive case of disseminated juvenile xanthogranuloma

Rikhia Chakraborty; Oliver A. Hampton; Harshal Abhyankar; Daniel Zinn; Amanda Grimes; Brooks Skull; Olive S. Eckstein; Nadia Mahmood; David A. Wheeler; Dolores Lopez-Terrada; Tricia L. Peters; John Hicks; Tarek Elghetany; Robert A. Krance; Poulikos I. Poulikakos; Miriam Merad; Kenneth L. McClain; Carl E. Allen; Williams D. Parsons

By controlling and limiting inflammatory conditions, naturally occurring regulatory T cells (Tregs), defined as circulating CD4+CD25brightFoxP3+ cells, play critical roles in maintaining tolerance and preventing autoimmunity and thus have tremendous potential for adoptive immunotherapy. Because they represent a scanty subset of the CD4+ T-lymphocyte subset, several approaches have been developed to isolate and expand ex vivo polyclonal Tregs. However, one limitation of the functional analyses performed on these cultured Tregs is the incomplete characterization of their tissue-trafficking properties. As this aspect provides crucial information for their therapeutic effects, we have here explored the chemokine receptor expression profile and function of Tregs cultured ex vivo with validated expansion protocols. Our data show that ex vivo cultured Tregs retained the expression of CCR7 but dramatically downregulated CCR5 as compared with freshly isolated Tregs. The differential chemokine receptors expression pattern corroborated with their respective steady state messenger RNA expression and also with their migration toward specific chemokines. Our analyses suggest that ex vivo cultured Tregs may display impaired or suboptimal migration to the inflamed tissues releasing RANTES and MIP-1&agr; chemokines.


Journal of Experimental Medicine | 2018

RAF/MEK/extracellular signal–related kinase pathway suppresses dendritic cell migration and traps dendritic cells in Langerhans cell histiocytosis lesions

Brandon Hogstad; Marie-Luise Berres; Rikhia Chakraborty; Jun Tang; Camille Bigenwald; Madhavika N. Serasinghe; Karen Phaik Har Lim; Howard Lin; Tsz-Kwong Man; Romain Remark; Samantha Baxter; Veronika Kana; Stefan Jordan; Zoi Karoulia; Wing-hong Kwan; Marylene Leboeuf; Elisa Brandt; Hélène Salmon; Kenneth L. McClain; Poulikos I. Poulikakos; Jerry E. Chipuk; Willem J. M. Mulder; Carl E. Allen; Miriam Merad

Juvenile xanthogranuloma (JXG) is a rare histiocytic disorder that is usually benign and self-limiting. We present a case of atypical, aggressive JXG harboring a novel mitogen-activated protein kinase (MAPK) pathway mutation in the MAPK1 gene, which encodes mitogen-activated protein kinase 1 or extracellular signal-regulated 2 (ERK2). Our analysis revealed that the mutation results in constitutive ERK activation that is resistant to BRAF or MEK inhibitors but susceptible to an ERK inhibitor. These data highlight the importance of identifying specific MAPK pathway alterations as part of the diagnostic workup for patients with histiocytic disorders rather than initiating empiric treatment with MEK inhibitors.


Clinical Cancer Research | 2017

p53 Nongenotoxic Activation and mTORC1 Inhibition Lead to Effective Combination for Neuroblastoma Therapy

Myrthala Moreno-Smith; Anna Lakoma; Zaowen Chen; Ling Tao; Kathleen A. Scorsone; Linda Schild; Kevin Aviles-Padilla; Rana Nikzad; Yankai Zhang; Rikhia Chakraborty; Jan J. Molenaar; Sanjeev A. Vasudevan; Vivien Sheehan; Eugene S. Kim; Silke Paust; Jason M. Shohet; Eveline Barbieri

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with constitutively activated mitogen-activated protein kinase (MAPK) pathway signaling. Approximately 60% of LCH patients harbor somatic BRAFV600E mutations localizing to CD207+ DCs within lesions. However, the mechanisms driving BRAFV600E+ LCH cell accumulation in lesions remain unknown. Here we show that sustained extracellular signal–related kinase activity induced by BRAFV600E inhibits C-C motif chemokine receptor 7 (CCR7)–mediated DC migration, trapping DCs in tissue lesions. Additionally, BRAFV600E increases expression of BCL2-like protein 1 (BCL2L1) in DCs, resulting in resistance to apoptosis. Pharmacological MAPK inhibition restores migration and apoptosis potential in a mouse LCH model, as well as in primary human LCH cells. We also demonstrate that MEK inhibitor-loaded nanoparticles have the capacity to concentrate drug delivery to phagocytic cells, significantly reducing off-target toxicity. Collectively, our results indicate that MAPK tightly suppresses DC migration and augments DC survival, rendering DCs in LCH lesions trapped and resistant to cell death.


Blood | 2017

A genome-wide association study of LCH identifies a variant in SMAD6 associated with susceptibility

Erin C. Peckham-Gregory; Rikhia Chakraborty; Michael E. Scheurer; John W. Belmont; Harshal Abhyankar; Amel G. Sengal; Brooks Scull; Olive S. Eckstein; Daniel Zinn; Louisa Mayer; Albert Shih; Miriam Merad; D. Williams Parsons; Kenneth L. McClain; Philip J. Lupo; Carl E. Allen

Purpose: mTORC1 inhibitors are promising agents for neuroblastoma therapy; however, they have shown limited clinical activity as monotherapy, thus rational drug combinations need to be explored to improve efficacy. Importantly, neuroblastoma maintains both an active p53 and an aberrant mTOR signaling. Experimental Design: Using an orthotopic xenograft model and modulating p53 levels, we investigated the antitumor effects of the mTORC1 inhibitor temsirolimus in neuroblastoma expressing normal, decreased, or mutant p53, both as single agent and in combination with first- and second-generation MDM2 inhibitors to reactivate p53. Results: Nongenotoxic p53 activation suppresses mTOR activity. Moreover, p53 reactivation via RG7388, a second-generation MDM2 inhibitor, strongly enhances the in vivo antitumor activity of temsirolimus. Single-agent temsirolimus does not elicit apoptosis, and tumors rapidly regrow after treatment suspension. In contrast, our combination therapy triggers a potent apoptotic response in wild-type p53 xenografts and efficiently blocks tumor regrowth after treatment completion. We also found that this combination uniquely led to p53-dependent suppression of survivin whose ectopic expression is sufficient to rescue the apoptosis induced by our combination. Conclusions: Our study supports a novel highly effective strategy that combines RG7388 and temsirolimus in wild-type p53 neuroblastoma, which warrants testing in early-phase clinical trials. Clin Cancer Res; 23(21); 6629–39. ©2017 AACR.

Collaboration


Dive into the Rikhia Chakraborty's collaboration.

Top Co-Authors

Avatar

Carl E. Allen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harshal Abhyankar

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Albert Shih

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Brooks Scull

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Daniel Zinn

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olive S. Eckstein

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Howard Lin

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge