Network


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

Hotspot


Dive into the research topics where Anuk Das is active.

Publication


Featured researches published by Anuk Das.


The Journal of Allergy and Clinical Immunology | 2008

Surfactant protein D inhibits TNF-α production by macrophages and dendritic cells in mice

L. Hortobagyi; S. Kierstein; Kateryna Krytska; Xiaoping Zhu; Anuk Das; Francis R. Poulain; Angela Haczku

BACKGROUNDnSurfactant protein (SP) D shares target cells with the proinflammatory cytokine TNF-alpha, an important autocrine stimulator of dendritic cells and macrophages in the airways.nnnOBJECTIVEnWe sought to study the mechanisms by which TNF-alpha and SP-D can affect cellular components of the pulmonary innate immune system.nnnMETHODSnCytokine and SP-D protein and mRNA expression was assessed by means of ELISA, Western blotting, and real-time PCR, respectively, by using in vivo models of allergic airway sensitization. Macrophage and dendritic cell phenotypes were analyzed by means of FACS analysis. Maturation of bone marrow-derived dendritic cells was investigated in vitro.nnnRESULTSnTNF-alpha, elicited either by allergen exposure or pulmonary overexpression, induced SP-D, IL-13, and mononuclear cell influx in the lung. Recombinant IL-13 by itself was also capable of enhancing SP-D in vivo and in vitro, and the SP-D response to allergen challenge was impaired in IL-13-deficient mice. Allergen-induced increase of SP-D in the airways coincided with resolution of TNF-alpha release and cell influx. SP-D-deficient mice had constitutively high numbers of alveolar mononuclear cells expressing TNF-alpha, MHC class II, CD86, and CD11b, characteristics of proinflammatory, myeloid dendritic cells. Recombinant SP-D significantly suppressed all of these molecules in bone marrow-derived dendritic cell cultures.nnnCONCLUSIONSnTNF-alpha can contribute to enhanced SP-D production in the lung indirectly through inducing IL-13. SP-D, on the other hand, can antagonize the proinflammatory effects of TNF-alpha on macrophages and dendritic cells, at least partly, by inhibiting production of this cytokine.


American Journal of Pathology | 2011

Targeting ST2L Potentiates CpG-Mediated Therapeutic Effects in a Chronic Fungal Asthma Model

Hemanth Ramaprakash; Takehiko Shibata; Karen E. Duffy; Ugur B. Ismailoglu; Rachel M. Bredernitz; Ana Paula Moreira; Ana Lucia Coelho; Anuk Das; Natalie Fursov; Geoffrey L. Chupp; Cory M. Hogaboam

IL-33 and its soluble receptor and cell-associated receptor (ST2L) are all increased in clinical and experimental asthma. The present study addressed the hypothesis that ST2L impairs the therapeutic effects of CpG in a fungal model of asthma. C57BL/6 mice were sensitized to Aspergillus fumigatus and challenged via i.t. instillation with live A. fumigatus conidia. Mice were treated with IgG alone, anti-ST2L monoclonal antibody (mAb) alone, CpG alone, IgG plus CpG, or anti-ST2L mAb plus CpG every other day from day 14 to day 28 and investigated on day 28 after conidia. Lung ST2L and toll-like receptor 9 protein expression levels concomitantly increased in a time-dependent manner during fungal asthma. Therapeutic blockade of ST2L with an mAb attenuated key pathological features of this model. At subtherapeutic doses, neither anti-ST2L mAb nor CpG alone affected fungal asthma severity. However, airway hyperresponsiveness, mucus cell metaplasia, peribronchial fibrosis, and fungus retention were markedly reduced in asthmatic mice treated with the combination of both. Whole lung CXCL9 levels were significantly elevated in the combination group but not in the controls. Furthermore, in asthmatic mice treated with the combination therapy, dendritic cells generated significantly greater IL-12p70 with CpG in vitro compared with control dendritic cells. The combination of anti-ST2L mAb with CpG significantly attenuated experimental asthma, suggesting that targeting ST2L might enhance the therapeutic efficacy of CpG during allergic inflammation.


Respiratory Research | 2015

Asthma characteristics and biomarkers from the Airways Disease Endotyping for Personalized Therapeutics (ADEPT) longitudinal profiling study

Philip E. Silkoff; I. Strambu; Michel Laviolette; Dave Singh; J. M. FitzGerald; Stephen Lam; Steven G. Kelsen; A. Eich; A. Ludwig-Sengpiel; G. C hupp; Vibeke Backer; Celeste Porsbjerg; P. O. Girodet; P. Berger; Richard Leigh; Joel N. Kline; Mark T. Dransfield; William J. Calhoun; Azra Hussaini; Sumita Khatri; Pascal Chanez; Vedrana S. Susulic; Elliot S. Barnathan; Mark E. Curran; Anuk Das; Carrie Brodmerkel; Frédéric Baribaud; Matthew J. Loza

BackgroundAsthma is a heterogeneous disease and development of novel therapeutics requires an understanding of pathophysiologic phenotypes. The purpose of the ADEPT study was to correlate clinical features and biomarkers with molecular characteristics, by profiling asthma (NCT01274507). This report presents for the first time the study design, and characteristics of the recruited subjects.MethodsPatients with a range of asthma severity and healthy non-atopic controls were enrolled. The asthmatic subjects were followed for 12xa0months. Assessments included history, patient questionnaires, spirometry, airway hyper-responsiveness to methacholine, fractional exhaled nitric oxide (FENO), and biomarkers measured in induced sputum, blood, and bronchoscopy samples. All subjects underwent sputum induction and 30 subjects/cohort had bronchoscopy.ResultsMild (nu2009=u200952), moderate (nu2009=u200955), severe (nu2009=u200951) asthma cohorts and 30 healthy controls were enrolled from North America and Western Europe. Airflow obstruction, bronchodilator response and airways hyperresponsiveness increased with asthma severity, and severe asthma subjects had reduced forced vital capacity. Asthma control questionnaire-7 (ACQ7) scores worsened with asthma severity. In the asthmatics, mean values for all clinical and biomarker characteristics were stable over 12xa0months although individual variability was evident. FENO and blood eosinophils did not differ by asthma severity. Induced sputum eosinophils but not neutrophils were lower in mild compared to the moderate and severe asthma cohorts.ConclusionsThe ADEPT study successfully enrolled asthmatics across a spectrum of severity and non-atopic controls. Clinical characteristics were related to asthma severity and in general asthma characteristics e.g. lung function, were stable over 12xa0months. Use of the ADEPT data should prove useful in defining biological phenotypes to facilitate personalized therapeutic approaches.


International Journal of Toxicology | 2008

Preclinical safety and pharmacology of an anti-human interleukin-13 monoclonal antibody in normal macaques and in macaques with allergic asthma.

Pauline L. Martin; Dusti Fisher; William Glass; Karyn O’Neil; Anuk Das; Elise Martin; Li Li

Interleukin-13 (IL-13) plays a central role in chronic airway diseases, including asthma. These studies were conducted to evaluate the safety of administration of a human anti-IL-13 monoclonal antibody (mAb) to normal macaques and in macaques with allergic asthma. In addition, serum and bronchioalveolar lavage fluid were collected from allergic cynomolgus macaques in order to identify potential surrogate markers of IL-13 pharmacology that could be useful for subsequent clinical trials. In vitro studies demonstrated that the anti-IL-13 mAb inhibited the pharmacological actions of both human and cynomolgus macaque IL-13. Allergic macaques were treated systemically with 10 mg/kg anti-IL-13 mAb 1 day prior to inhaled Ascaris suum antigen challenge. Normal macaques were dosed intravenously with anti-IL-13 once per week for 3 weeks at doses of 10 or 50 mg/kg. Treatment of macaques with the anti-IL-13 mAb was not associated with any toxicologically significant findings. A slight treatment-related but nonadverse decrease in platelet counts was observed in both the normal and allergic macaques. In allergic macaques, the anti-IL-13 mAb treatment did not affect lung function, lung eosinophilia, or serum or BAL immunoglobulin E (IgE) concentrations but did produce a reduction in BAL and serum eotaxin concentrations ( p < .05) at 6 h post antigen challenge. This study shows that administration of an anti-IL-13 mAb was well tolerated in both normal and allergic asthmatic macaques and that serum eotaxin concentrations may be a useful early in vivo marker for evaluating IL-13 inhibition in patients with asthma.


Nature Genetics | 2017

A functional genomics predictive network model identifies regulators of inflammatory bowel disease

Lauren A. Peters; Jacqueline Perrigoue; Arthur Mortha; Alina C. Iuga; Won Min Song; Eric M. Neiman; Sean R. Llewellyn; Antonio Di Narzo; Brian A. Kidd; Shannon Telesco; Yongzhong Zhao; Aleksandar Stojmirovic; Jocelyn Sendecki; Khader Shameer; Riccardo Miotto; Bojan Losic; Hardik Shah; Eunjee Lee; Minghui Wang; Jeremiah J. Faith; Andrew Kasarskis; Carrie Brodmerkel; Mark E. Curran; Anuk Das; Joshua R. Friedman; Yoshinori Fukui; Mary Beth Humphrey; Brian M. Iritani; Nicholas Sibinga; Teresa K. Tarrant

A major challenge in inflammatory bowel disease (IBD) is the integration of diverse IBD data sets to construct predictive models of IBD. We present a predictive model of the immune component of IBD that informs causal relationships among loci previously linked to IBD through genome-wide association studies (GWAS) using functional and regulatory annotations that relate to the cells, tissues, and pathophysiology of IBD. Our model consists of individual networks constructed using molecular data generated from intestinal samples isolated from three populations of patients with IBD at different stages of disease. We performed key driver analysis to identify genes predicted to modulate network regulatory states associated with IBD, prioritizing and prospectively validating 12 of the top key drivers experimentally. This validated key driver set not only introduces new regulators of processes central to IBD but also provides the integrated circuits of genetic, molecular, and clinical traits that can be directly queried to interrogate and refine the regulatory framework defining IBD.


The Journal of Allergy and Clinical Immunology | 2017

Activated plasmacytoid dendritic cells regulate type 2 innate lymphoid cell–mediated airway hyperreactivity

Hadi Maazi; Homayon Banie; German R. Aleman Muench; Nisheel Patel; Bowen Wang; Ishwarya Sankaranarayanan; Vipul Bhargava; Takahiro Sato; Gavin Lewis; Matteo Cesaroni; James Karras; Anuk Das; Pejman Soroosh; Omid Akbari

Background: Allergic asthma is a prevalent inflammatory disease of the airways caused by dysregulated immune balance in the lungs with incompletely understood pathogenesis. The recently identified type 2 innate lymphoid cells (ILC2s) play significant roles in the pathogenesis of asthma. Although ILC2‐activating factors have been identified, the mechanisms that suppress ILC2s remain largely unknown. Plasmacytoid dendritic cells (pDCs) are important in antiviral immunity and in maintaining tolerance to inert antigens. Objective: We sought to address the role of pDCs in regulating ILC2 function and ILC2‐mediated airway hyperreactivity (AHR) and lung inflammation. Methods: We used several murine models, including BDCA‐2–diphtheria toxin receptor (DTR) transgenic and IFN‐&agr; receptor 1–deficient mice, as well as purified primary ILC2s, to reach our objective. We extended and validated our findings to human ILC2s. Results: We show that activation of pDCs through Toll‐like receptor 7/8 suppresses ILC2‐mediated AHR and airway inflammation and that depletion of pDCs reverses this suppression. We further show that pDCs suppress cytokine production and the proliferation rate while increasing the apoptosis rate of ILC2s through IFN‐&agr; production. Transcriptomic analysis of both human and murine ILC2s confirms the activation of regulatory pathways in ILC2s by IFN‐&agr;. Conclusion: Activation of pDCs alleviates AHR and airway inflammation by suppressing ILC2 function and survival. Our findings reveal a novel regulatory pathway in ILC2‐mediated pulmonary inflammation with important clinical implications.


Current Pharmaceutical Design | 2006

Biopharmaceutical therapeutics for asthma remodeling.

Anuk Das; Don E. Griswold; Theodore J. Torphy; Li Li

Current asthma therapy is aimed at controlling disease symptoms. A subset of asthma patients remains symptomatic despite optimal therapy indicating that an unmet medical need exists for these patients. Innovative therapeutics are needed to treat the unmet need in asthma and biopharmaceutical approaches may provide an opportunity for identifying these agents. It is proposed that airway remodeling contributes to asthma symptoms and this feature of disease pathology may be a target for future therapies. The current review focuses on the contribution of one feature of airway remodeling, subepithelial fibrosis, towards disease and highlights some of the mechanisms that may contribute to this feature of airway remodeling. Further, some potential molecular targets are identified for consideration for therapeutic intervention.


bioRxiv | 2018

Inflammatory bowel disease microbiotas alter gut CD4 T-cell homeostasis and drive colitis in mice

Graham J Britton; Eduardo J Contijoch; Ilaria Mogno; Olivia H. Vennaro; Sean R. Llewellyn; Ruby Ng; Zhihua Li; Arthur Mortha; Miriam Merad; Anuk Das; Dirk Gevers; Dermot P. McGovern; Namita Singh; Jonathan Braun; Jonathan P. Jacobs; Jose C. Clemente; Ari Grinspan; Bruce E. Sands; Jean-Frederic Colombel; Marla C. Dubinsky; Jeremiah J. Faith

To examine the functional contribution of Inflammatory Bowel Disease (IBD) microbes to immune homeostasis and colitis, we colonized unchallenged and colitis-susceptible germ-free mice with over twenty human intestinal microbiotas from healthy and IBD donors. Compared to healthy microbiotas, IBD microbiotas led to expanded RORγt+Th17 cells and reduced RORγt+Treg in the gut of unchallenged gnotobiotic mice and increased disease severity in colitis-susceptible mice. The proportions of RORγt+Th17 and RORγt+Treg induced by each microbiota were highly predictive of the human disease status and strongly correlated with disease severity in colitis-susceptible mice colonized with the same human microbiotas. The transmittable functional potential of IBD microbes suggests a mechanism for a microbial contribution to IBD pathogenesis and a potential route for its treatment and prevention.


bioRxiv | 2018

A negative role for the interleukin-2-inducible T-cell kinase (ITK) in human Foxp3+ Treg differentiation

Polina Mamontov; Ryan A Eberwine; Jackie Perrigoue; Anuk Das; Joshua R. Friedman; Jorge R Mora

The Tec kinases ITK (interleukin-2-inducible T-cell kinase) and RLK (resting lymphocyte kinase) are critical components of the proximal TCR/CD3 signal transduction machinery, and data in mice suggest that ITK negatively regulates TREG differentiation. However, whether Tec kinases modulate TREG development and/or function in human T cells remains unknown. Using a novel self-delivery siRNA platform (sdRNA), we found that ITK knockdown in primary human naïve peripheral blood CD4 T cells increased Foxp3+ TREG differentiation under both TREG and T effector (Teff) cell priming conditions. ITK knockdown also enhanced the expression of the co-inhibitory receptor PD-1 on FoxP3+ T cells. TREGS differentiated in vitro (iTREG) after ITK knockdown displayed suppressive capacity against effector CD4+ T cell proliferation. ITK knockdown decreased IL-17A production in T cells primed under Th17 conditions and increased Th1 differentiation. Finally, a dual ITK/RLK Tec kinase inhibitor blocked TREG differentiation and T cell activation in general. Our data suggest that targeting ITK in human T cells may be an effective approach to boost TREG in the context of autoimmune diseases, but non-specific inhibition of other Tec family kinases may broadly inhibit T cell activation.


Cytokine | 2004

Anti-IL-13 monoclonal antibody inhibits airway hyperresponsiveness, inflammation and airway remodeling.

Gaoyun Yang; Amy Volk; Ted Petley; Eva Emmell; Jill Giles-Komar; Xiaozhou Shang; Jian Li; Anuk Das; Dave Shealy; Don E. Griswold; Li Li

Collaboration


Dive into the Anuk Das's collaboration.

Top Co-Authors

Avatar

Li Li

Janssen Pharmaceutica

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angela Haczku

University of California

View shared research outputs
Top Co-Authors

Avatar

Arthur Mortha

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge