Network


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

Hotspot


Dive into the research topics where Dawn Chapelle is active.

Publication


Featured researches published by Dawn Chapelle.


The New England Journal of Medicine | 2009

An Autoinflammatory Disease with Deficiency of the Interleukin-1–Receptor Antagonist

Ivona Aksentijevich; Seth L. Masters; Polly J. Ferguson; Paul Dancey; Joost Frenkel; Annet van Royen-Kerkhoff; Ron Laxer; Ulf Tedgård; Edward W. Cowen; Tuyet-Hang Pham; Matthew G. Booty; Jacob D. Estes; Netanya G. Sandler; Nicole Plass; Deborah L. Stone; Maria L. Turner; Suvimol Hill; Rayfel Schneider; Paul Babyn; Hatem El-Shanti; Elena Pope; Karyl S. Barron; Xinyu Bing; Arian Laurence; Chyi-Chia R. Lee; Dawn Chapelle; Gillian I. Clarke; Kamal Ohson; Marc Nicholson; Massimo Gadina

BACKGROUND Autoinflammatory diseases manifest inflammation without evidence of infection, high-titer autoantibodies, or autoreactive T cells. We report a disorder caused by mutations of IL1RN, which encodes the interleukin-1-receptor antagonist, with prominent involvement of skin and bone. METHODS We studied nine children from six families who had neonatal onset of sterile multifocal osteomyelitis, periostitis, and pustulosis. Response to empirical treatment with the recombinant interleukin-1-receptor antagonist anakinra in the first patient prompted us to test for the presence of mutations and changes in proteins and their function in interleukin-1-pathway genes including IL1RN. RESULTS We identified homozygous mutations of IL1RN in nine affected children, from one family from Newfoundland, Canada, three families from The Netherlands, and one consanguineous family from Lebanon. A nonconsanguineous patient from Puerto Rico was homozygous for a genomic deletion that includes IL1RN and five other interleukin-1-family members. At least three of the mutations are founder mutations; heterozygous carriers were asymptomatic, with no cytokine abnormalities in vitro. The IL1RN mutations resulted in a truncated protein that is not secreted, thereby rendering cells hyperresponsive to interleukin-1beta stimulation. Patients treated with anakinra responded rapidly. CONCLUSIONS We propose the term deficiency of the interleukin-1-receptor antagonist, or DIRA, to denote this autosomal recessive autoinflammatory disease caused by mutations affecting IL1RN. The absence of interleukin-1-receptor antagonist allows unopposed action of interleukin-1, resulting in life-threatening systemic inflammation with skin and bone involvement. (ClinicalTrials.gov number, NCT00059748.)


Nature Genetics | 2014

An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome

Scott W. Canna; Adriana A. Jesus; Sushanth Gouni; Stephen R. Brooks; Bernadette Marrero; Yin Liu; Michael A. DiMattia; Kristien J M Zaal; Gina A. Montealegre Sanchez; Hanna Kim; Dawn Chapelle; Nicole Plass; Yan Huang; Alejandro V. Villarino; Angélique Biancotto; Thomas A. Fleisher; Joseph A. Duncan; John J. O'Shea; Susanne M. Benseler; Alexei A. Grom; Zuoming Deng; Ronald M. Laxer; Raphaela Goldbach-Mansky

Inflammasomes are innate immune sensors that respond to pathogen- and damage-associated signals with caspase-1 activation, interleukin (IL)-1β and IL-18 secretion, and macrophage pyroptosis. The discovery that dominant gain-of-function mutations in NLRP3 cause the cryopyrin-associated periodic syndromes (CAPS) and trigger spontaneous inflammasome activation and IL-1β oversecretion led to successful treatment with IL-1–blocking agents. Herein we report a de novo missense mutation (c.1009A>T, encoding p.Thr337Ser) affecting the nucleotide-binding domain of the inflammasome component NLRC4 that causes early-onset recurrent fever flares and macrophage activation syndrome (MAS). Functional analyses demonstrated spontaneous inflammasome formation and production of the inflammasome-dependent cytokines IL-1β and IL-18, with the latter exceeding the levels seen in CAPS. The NLRC4 mutation caused constitutive caspase-1 cleavage in cells transduced with mutant NLRC4 and increased production of IL-18 in both patient-derived and mutant NLRC4–transduced macrophages. Thus, we describe a new monoallelic inflammasome defect that expands the monogenic autoinflammatory disease spectrum to include MAS and suggests new targets for therapy.


Arthritis & Rheumatism | 2012

Sustained Response and Prevention of Damage Progression in Patients With Neonatal-Onset Multisystem Inflammatory Disease Treated With Anakinra: A Cohort Study to Determine Three- and Five-Year Outcomes

Cailin Sibley; Nikki Plass; Joseph Snow; Edythe Wiggs; Carmen C. Brewer; Kelly A. King; Christopher Zalewski; H. Jeffrey Kim; Rachel J. Bishop; Suvimol Hill; Scott M. Paul; Patrick Kicker; Zachary Phillips; Joseph G. Dolan; Brigitte C. Widemann; Nalini Jayaprakash; Frank Pucino; Deborah L. Stone; Dawn Chapelle; Christopher Snyder; Robert Wesley; Raphaela Goldbach-Mansky

OBJECTIVE Blocking interleukin-1 with anakinra in patients with the autoinflammatory syndrome neonatal-onset multisystem inflammatory disease (NOMID) reduces systemic and organ-specific inflammation. However, the impact of long-term treatment has not been established. This study was undertaken to evaluate the long-term effect of anakinra on clinical and laboratory outcomes and safety in patients with NOMID. METHODS We conducted a cohort study of 26 NOMID patients ages 0.80-42.17 years who were followed up at the NIH and treated with anakinra 1-5 mg/kg/day for at least 36 months. Disease activity was assessed using daily diaries, questionnaires, and C-reactive protein level. Central nervous system (CNS) inflammation, hearing, vision, and safety were evaluated. RESULTS Sustained improvements in diary scores, parents/patients and physicians global scores of disease activity, parents/patients pain scores, and inflammatory markers were observed (all P<0.001 at 36 and 60 months). At 36 and 60 months, CNS inflammation was suppressed, with decreased cerebrospinal fluid white blood cell counts (P=0.0026 and P=0.0076, respectively), albumin levels, and opening pressures (P=0.0012 and P<0.001, respectively). Most patients showed stable or improved hearing. Cochlear enhancement on magnetic resonance imaging correlated with continued hearing loss. Visual acuity and peripheral vision were stable. Low optic nerve size correlated with poor visual field. Bony lesions progressed. Adverse events other than viral infections were rare, and all patients continued to receive the medication. CONCLUSION These findings indicate that anakinra provides sustained efficacy in the treatment of NOMID for up to 5 years, with the requirement of dose escalation. Damage progression in the CNS, ear, and eye, but not bone, is preventable. Anakinra is well tolerated overall.


Journal of Clinical Investigation | 2015

Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production

Anja Brehm; Yin Liu; Afzal Sheikh; Bernadette Marrero; Ebun Omoyinmi; Qing Zhou; Gina Montealegre; Angélique Biancotto; Adam Reinhardt; Adriana A. Jesus; Martin Pelletier; Wanxia L. Tsai; Elaine F. Remmers; Lela Kardava; Suvimol Hill; Hanna Kim; Helen J. Lachmann; André Mégarbané; Jae Jin Chae; Jilian Brady; Rhina D. Castillo; Diane Brown; Angel Vera Casano; Ling Gao; Dawn Chapelle; Yan Huang; Deborah L. Stone; Yongqing Chen; Franziska Sotzny; Chyi-Chia Richard Lee

Autosomal recessive mutations in proteasome subunit β 8 (PSMB8), which encodes the inducible proteasome subunit β5i, cause the immune-dysregulatory disease chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE), which is classified as a proteasome-associated autoinflammatory syndrome (PRAAS). Here, we identified 8 mutations in 4 proteasome genes, PSMA3 (encodes α7), PSMB4 (encodes β7), PSMB9 (encodes β1i), and proteasome maturation protein (POMP), that have not been previously associated with disease and 1 mutation in PSMB8 that has not been previously reported. One patient was compound heterozygous for PSMB4 mutations, 6 patients from 4 families were heterozygous for a missense mutation in 1 inducible proteasome subunit and a mutation in a constitutive proteasome subunit, and 1 patient was heterozygous for a POMP mutation, thus establishing a digenic and autosomal dominant inheritance pattern of PRAAS. Function evaluation revealed that these mutations variably affect transcription, protein expression, protein folding, proteasome assembly, and, ultimately, proteasome activity. Moreover, defects in proteasome formation and function were recapitulated by siRNA-mediated knockdown of the respective subunits in primary fibroblasts from healthy individuals. Patient-isolated hematopoietic and nonhematopoietic cells exhibited a strong IFN gene-expression signature, irrespective of genotype. Additionally, chemical proteasome inhibition or progressive depletion of proteasome subunit gene transcription with siRNA induced transcription of type I IFN genes in healthy control cells. Our results provide further insight into CANDLE genetics and link global proteasome dysfunction to increased type I IFN production.


Annals of the Rheumatic Diseases | 2015

A 24-month open-label study of canakinumab in neonatal-onset multisystem inflammatory disease

Cailin Sibley; Andrea Chioato; Sd Felix; Laurence Colin; Abhijit Chakraborty; Nikki Plass; Jackeline Rodriguez-Smith; Carmen C. Brewer; Kelly A. King; Christopher Zalewski; H. Jeffrey Kim; Rachel J. Bishop; Ken Abrams; Deborah L. Stone; Dawn Chapelle; Bahar Kost; Christopher Snyder; Robert Wesley; Raphaela Goldbach-Mansky

Objective To study efficacy and safety of escalating doses of canakinumab, a fully human anti-IL-1β monoclonal antibody in the severe cryopyrin-associated periodic syndrome, neonatal-onset multisystem inflammatory disease (NOMID). Methods 6 patients were enrolled in this 24-month, open-label phase I/II study. All underwent anakinra withdrawal. The initial subcutaneous canakinumab dose was 150 mg (or 2 mg/kg in patients ≤40 kg) or 300 mg (or 4 mg/kg) with escalation up to 600 mg (or 8 mg/kg) every 4 weeks. Full remission was remission of patient-reported clinical components and measures of systemic inflammation and CNS inflammation. Hearing, vision and safety were assessed. Primary endpoint was full remission at month 6. Results All patients flared after anakinra withdrawal, and symptoms and serum inflammatory markers improved with canakinumab. All patients required dose escalation to the maximum dose. At month 6, none had full remission, although 4/6 achieved inflammatory remission, based on disease activity diary scores and normal C-reactive proteins. None had CNS remission; 5/6 due to persistent CNS leucocytosis. At the last study visit, 5/6 patients achieved inflammatory remission and 4/6 had continued CNS leucocytosis. Visual acuity and field were stable in all patients, progressive hearing loss occurred in 1/10 ears. Adverse events (AEs) were rare. One serious AE (abscess due to a methicillin-resistant Staphylococcus aureus infection) occurred. Conclusions Canakinumab at the studied doses improves symptoms and serum inflammatory features of NOMID, although low-grade CNS leukocytosis in four patients and headaches in one additional patient persisted. Whether further dose intensifications are beneficial in these cases remains to be assessed. ClinicalTrials.gov identifier NCT00770601.


Arthritis & Rheumatism | 2014

Anakinra use during pregnancy in patients with cryopyrin-associated periodic syndromes (CAPS).

Zenas Chang; Catherine Y. Spong; Adriana A. Jesus; Michael A. Davis; Nicole Plass; Deborah L. Stone; Dawn Chapelle; Patrycja Hoffmann; Daniel L. Kastner; Karyl S. Barron; Raphaela Goldbach-Mansky; Pamela Stratton

To describe the pregnancy course and outcome and the use of anakinra, a recombinant selective interleukin‐1 receptor blocker, during pregnancy in patients with cryopyrin‐associated periodic syndromes (CAPS), including familial cold autoinflammatory syndrome (FCAS), Muckle‐Wells syndrome (MWS), and neonatal‐onset multisystem inflammatory disease (NOMID).


Pediatric Rheumatology | 2015

Preliminary response to Janus kinase inhibition with baricitinib in chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperatures (CANDLE)

Gina Montealegre; Adam Reinhardt; Paul A. Brogan; Yy Berkun; Abraham Zlotogorski; D. Brown; P. Chira; Ling Gao; Jason A. Dare; S. Schalm; R. Merino; Dawn Chapelle; Hanna Kim; S. Judd; M. O'Brien; A Almeida de Jesus; Y. Kim; B. Kost; Yan Huang; S.M. Paul; A. Brofferio; Chyi Chia Richard Lee; C. Hadigan; T. Heller; C. Minniti; Kristina I. Rother; Raphaela Goldbach-Mansky

Preliminary response to Janus kinase inhibition with baricitinib in chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperatures (CANDLE) G Montealegre, A Reinhardt, P Brogan, Y Berkun, A Zlotogorski, D Brown, P Chira, L Gao, J Dare, S Schalm, R Merino, D Chapelle, H Kim, S Judd, M O’Brien, A Almeida De Jesus, Y Kim, B Kost, Y Huang, S Paul, A Brofferio, C-C Lee, C Hadigan, T Heller, C Minniti, K Rother, R Goldbach-Mansky


Arthritis & Rheumatism | 2017

Cerebrospinal Fluid Cytokines Correlate With Aseptic Meningitis and Blood–Brain Barrier Function in Neonatal-Onset Multisystem Inflammatory Disease: Central Nervous System Biomarkers in Neonatal-Onset Multisystem Inflammatory Disease Correlate With Central Nervous System Inflammation

Jackeline Rodriguez-Smith; Yen Chih Lin; Wanxia Li Tsai; Hanna Kim; Gina Montealegre-Sanchez; Dawn Chapelle; Yan Huang; Cailin H. Sibley; Massimo Gadina; Robert Wesley; Bibiana Bielekova; Raphaela Goldbach-Mansky

To evaluate proinflammatory cytokines and leukocyte subpopulations in the cerebrospinal fluid (CSF) and blood of patients with neonatal‐onset multisystem inflammatory disease (NOMID) after treatment, and to compare inflammatory cytokines in the CSF and blood in 6 patients treated with 2 interleukin‐1 (IL‐1) blockers—anakinra and canakinumab.


Arthritis & Rheumatism | 2017

CSF cytokines correlate with aseptic meningitis and blood brain barrier function in Neonatal‐Onset Multisystem Inflammatory Disease (NOMID)

Jackeline Rodriguez-Smith; Yen-Chih Lin; Wanxia Li Tsai; Hanna Kim; Gina Montealegre-Sanchez; Dawn Chapelle; Yan Huang; Cailin Sibley; Massimo Gadina; Robert Wesley; Bibiana Bielekova; Raphaela Goldbach-Mansky

To evaluate proinflammatory cytokines and leukocyte subpopulations in the cerebrospinal fluid (CSF) and blood of patients with neonatal‐onset multisystem inflammatory disease (NOMID) after treatment, and to compare inflammatory cytokines in the CSF and blood in 6 patients treated with 2 interleukin‐1 (IL‐1) blockers—anakinra and canakinumab.


Journal of Clinical Investigation | 2016

Erratum: Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type i IFN production (Journal of Clinical Investigation (2015) 125:11 (4196-4211) DOI: 10.1172/JCI81260)

Anja Brehm; Yin Liu; Afzal Sheikh; Bernadette Marrero; Ebun Omoyinmi; Qing Zhou; Gina Montealegre; Angélique Biancotto; Adam Reinhardt; Adriana A. Jesus; Martin Pelletier; Wanxia L. Tsai; Elaine F. Remmers; Lela Kardava; Suvimol Hill; Hanna Kim; Helen J. Lachmann; André Mégarbané; Jae Jin Chae; Jilian Brady; Rhina D. Castillo; Diane Brown; Angel Vera Casano; Ling Gao; Dawn Chapelle; Yan Huang; Deborah L. Stone; Yongqing Chen; Franziska Sotzny; Chyi Chia Richard Lee

Autosomal recessive mutations in proteasome subunit b 8 (PSMB8), which encodes the inducible proteasome subunit b5i, cause the immune-dysregulatory disease chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE), which is classified as a proteasome-associated autoinflammatory syndrome (PRAAS). Here, we identified 8 mutations in 4 proteasome genes, PSMA3 (encodes a7), PSMB4 (encodes b7), PSMB9 (encodes b1i), and proteasome maturation protein (POMP), that have not been previously associated with disease and 1 mutation in PSMB8 that has not been previously reported. One patient was compound heterozygous for PSMB4 mutations, 6 patients from 4 families were heterozygous for a missense mutation in 1 inducible proteasome subunit and a mutation in a constitutive proteasome subunit, and 1 patient was heterozygous for a POMP mutation, thus establishing a digenic and autosomal dominant inheritance pattern of PRAAS. Function evaluation revealed that these mutations variably affect transcription, protein expression, protein folding, proteasome assembly, and, ultimately, proteasome activity. Moreover, defects in proteasome formation and function were recapitulated by siRNA-mediated knockdown of the respective subunits in primary fibroblasts from healthy individuals. Patient-isolated hematopoietic and nonhematopoietic cells exhibited a strong IFN gene-expression signature, irrespective of genotype. Additionally, chemical [...] Research Article Immunology

Collaboration


Dive into the Dawn Chapelle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Huang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hanna Kim

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Adriana A. Jesus

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Deborah L. Stone

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gina Montealegre

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicole Plass

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Bernadette Marrero

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Suvimol Hill

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yin Liu

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge