Network


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

Hotspot


Dive into the research topics where Joshua D. Milner is active.

Publication


Featured researches published by Joshua D. Milner.


Nature | 2008

Impaired T H 17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome

Joshua D. Milner; Jason M. Brenchley; Arian Laurence; Alexandra F. Freeman; Brenna J. Hill; Kevin M. Elias; Yuka Kanno; Christine Spalding; Houda Elloumi; Michelle L. Paulson; Joie Davis; Amy P. Hsu; Ava I. Asher; John J. O'Shea; Steven M. Holland; William E. Paul

The autosomal dominant hyper-IgE syndrome (HIES, ‘Job’s syndrome’) is characterized by recurrent and often severe pulmonary infections, pneumatoceles, eczema, staphylococcal abscesses, mucocutaneous candidiasis, and abnormalities of bone and connective tissue. Mutations presumed to underlie HIES have recently been identified in stat3, the gene encoding STAT3 (signal transducer and activator of transcription 3) (refs 3, 4). Although impaired production of interferon-γ and tumour-necrosis factor by T cells, diminished memory T-cell populations, decreased delayed-type-hypersensitivity responses and decreased in vitro lymphoproliferation in response to specific antigens have variably been described, specific immunological abnormalities that can explain the unique susceptibility to particular infections seen in HIES have not yet been defined. Here we show that interleukin (IL)-17 production by T cells is absent in HIES individuals. We observed that ex vivo T cells from subjects with HIES failed to produce IL-17, but not IL-2, tumour-necrosis factor or interferon-γ, on mitogenic stimulation with staphylococcal enterotoxin B or on antigenic stimulation with Candida albicans or streptokinase. Purified naive T cells were unable to differentiate into IL-17-producing (TH17) T helper cells in vitro and had lower expression of retinoid-related orphan receptor (ROR)-γt, which is consistent with a crucial role for STAT3 signalling in the generation of TH17 cells. TH17 cells have emerged as an important subset of helper T cells that are believed to be critical in the clearance of fungal and extracellular bacterial infections. Thus, our data suggest that the inability to produce TH17 cells is a mechanism underlying the susceptibility to the recurrent infections commonly seen in HIES.


The New England Journal of Medicine | 2014

Early-onset stroke and vasculopathy associated with mutations in ADA2

Qing Zhou; Dan Yang; Amanda K. Ombrello; Andrey Zavialov; Camilo Toro; Anton V. Zavialov; Deborah L. Stone; Jae Jin Chae; Sergio D. Rosenzweig; Kevin Bishop; Karyl S. Barron; Hye Sun Kuehn; Patrycja Hoffmann; Alejandra Negro; Wanxia L. Tsai; Edward W. Cowen; Wuhong Pei; Joshua D. Milner; Christopher Silvin; Theo Heller; David T. Chin; Nicholas J. Patronas; John S. Barber; Chyi-Chia R. Lee; Geryl Wood; Alexander Ling; Susan J. Kelly; David E. Kleiner; James C. Mullikin; Nancy J. Ganson

BACKGROUND We observed a syndrome of intermittent fevers, early-onset lacunar strokes and other neurovascular manifestations, livedoid rash, hepatosplenomegaly, and systemic vasculopathy in three unrelated patients. We suspected a genetic cause because the disorder presented in early childhood. METHODS We performed whole-exome sequencing in the initial three patients and their unaffected parents and candidate-gene sequencing in three patients with a similar phenotype, as well as two young siblings with polyarteritis nodosa and one patient with small-vessel vasculitis. Enzyme assays, immunoblotting, immunohistochemical testing, flow cytometry, and cytokine profiling were performed on samples from the patients. To study protein function, we used morpholino-mediated knockdowns in zebrafish and short hairpin RNA knockdowns in U937 cells cultured with human dermal endothelial cells. RESULTS All nine patients carried recessively inherited mutations in CECR1 (cat eye syndrome chromosome region, candidate 1), encoding adenosine deaminase 2 (ADA2), that were predicted to be deleterious; these mutations were rare or absent in healthy controls. Six patients were compound heterozygous for eight CECR1 mutations, whereas the three patients with polyarteritis nodosa or small-vessel vasculitis were homozygous for the p.Gly47Arg mutation. Patients had a marked reduction in the levels of ADA2 and ADA2-specific enzyme activity in the blood. Skin, liver, and brain biopsies revealed vasculopathic changes characterized by compromised endothelial integrity, endothelial cellular activation, and inflammation. Knockdown of a zebrafish ADA2 homologue caused intracranial hemorrhages and neutropenia - phenotypes that were prevented by coinjection with nonmutated (but not with mutated) human CECR1. Monocytes from patients induced damage in cocultured endothelial-cell layers. CONCLUSIONS Loss-of-function mutations in CECR1 were associated with a spectrum of vascular and inflammatory phenotypes, ranging from early-onset recurrent stroke to systemic vasculopathy or vasculitis. (Funded by the National Institutes of Health Intramural Research Programs and others.).


The New England Journal of Medicine | 2012

Cold Urticaria, Immunodeficiency, and Autoimmunity Related to PLCG2 Deletions

Michael J. Ombrello; Elaine F. Remmers; Guangping Sun; Alexandra F. Freeman; Shrimati Datta; Parizad Torabi-Parizi; Naeha Subramanian; Tom D. Bunney; Rhona W. Baxendale; Marta Martins; Neil Romberg; Hirsh D. Komarow; Ivona Aksentijevich; Hun Sik Kim; Jason Ho; Glenn Cruse; Mi-Yeon Jung; Alasdair M. Gilfillan; Dean D. Metcalfe; Celeste Nelson; Michelle O'Brien; Laura Wisch; Kelly D. Stone; Chhavi Gandhi; Alan A. Wanderer; Hane Lee; Stanley F. Nelson; Elizabeth T. Cirulli; David B. Goldstein; Eric O. Long

BACKGROUND Mendelian analysis of disorders of immune regulation can provide insight into molecular pathways associated with host defense and immune tolerance. METHODS We identified three families with a dominantly inherited complex of cold-induced urticaria, antibody deficiency, and susceptibility to infection and autoimmunity. Immunophenotyping methods included flow cytometry, analysis of serum immunoglobulins and autoantibodies, lymphocyte stimulation, and enzymatic assays. Genetic studies included linkage analysis, targeted Sanger sequencing, and next-generation whole-genome sequencing. RESULTS Cold urticaria occurred in all affected subjects. Other, variable manifestations included atopy, granulomatous rash, autoimmune thyroiditis, the presence of antinuclear antibodies, sinopulmonary infections, and common variable immunodeficiency. Levels of serum IgM and IgA and circulating natural killer cells and class-switched memory B cells were reduced. Linkage analysis showed a 7-Mb candidate interval on chromosome 16q in one family, overlapping by 3.5 Mb a disease-associated haplotype in a smaller family. This interval includes PLCG2, encoding phospholipase Cγ(2) (PLCγ(2)), a signaling molecule expressed in B cells, natural killer cells, and mast cells. Sequencing of complementary DNA revealed heterozygous transcripts lacking exon 19 in two families and lacking exons 20 through 22 in a third family. Genomic sequencing identified three distinct in-frame deletions that cosegregated with disease. These deletions, located within a region encoding an autoinhibitory domain, result in protein products with constitutive phospholipase activity. PLCG2-expressing cells had diminished cellular signaling at 37°C but enhanced signaling at subphysiologic temperatures. CONCLUSIONS Genomic deletions in PLCG2 cause gain of PLCγ(2) function, leading to signaling abnormalities in multiple leukocyte subsets and a phenotype encompassing both excessive and deficient immune function. (Funded by the National Institutes of Health Intramural Research Programs and others.).


Blood | 2015

Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations.

Joshua D. Milner; Tiphanie P. Vogel; Lisa R. Forbes; Chi A. Ma; Asbjørg Stray-Pedersen; Julie E. Niemela; Jonathan J. Lyons; Karin R. Engelhardt; Yu Zhang; Nermina Topcagic; Elisha D. O. Roberson; Helen F. Matthews; James W. Verbsky; Trivikram Dasu; Alexander Vargas-Hernández; Nidhy P. Varghese; Kenneth L. McClain; Lina Karam; Karen Nahmod; George Makedonas; Emily M. Mace; Hanne Sørmo Sorte; Gøri Perminow; V. Koneti Rao; Michael P. O’Connell; Susan Price; Helen C. Su; Morgan Butrick; Joshua McElwee; Jason D. Hughes

Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.


The Journal of Allergy and Clinical Immunology | 2013

Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation–polyendocrinopathy–enteropathy–X-linked–like syndrome

Gulbu Uzel; Elizabeth P. Sampaio; Monica G. Lawrence; Amy P. Hsu; Mary J. Hackett; Morna J. Dorsey; Richard J. Noel; James W. Verbsky; Alexandra F. Freeman; Erin Janssen; Francisco A. Bonilla; Joseph Pechacek; Prabha Chandrasekaran; Sarah K. Browne; Anahita Agharahimi; Ahmed M. Gharib; Sara Ciullini Mannurita; Jae Joon Yim; Eleonora Gambineri; Troy R. Torgerson; Dat Q. Tran; Joshua D. Milner; Steven M. Holland

BACKGROUND Mutations in signal transducer and activator of transcription (STAT) 1 cause a broad spectrum of disease, ranging from severe viral and bacterial infections (amorphic alleles) to mild disseminated mycobacterial disease (hypomorphic alleles) to chronic mucocutaneous candidiasis (CMC; hypermorphic alleles). The hypermorphic mutations are also associated with arterial aneurysms, autoimmunity, and squamous cell cancers. OBJECTIVE We sought to investigate the role of STAT1 gain-of-function mutations in phenotypes other than CMC. METHODS We initially screened patients with CMC and autoimmunity for STAT1 mutations. We functionally characterized mutations in vitro and studied immune profiles and regulatory T (Treg) cells. After our initial case identifications, we explored 2 large cohorts of patients with wild-type forkhead box protein 3 and an immune dysregulation-polyendocrinopathy-enteropathy-X-linked (IPEX)-like phenotype for STAT1 mutations. RESULTS We identified 5 children with polyendocrinopathy, enteropathy, and dermatitis reminiscent of IPEX syndrome; all but 1 had a variety of mucosal and disseminated fungal infections. All patients lacked forkhead box protein 3 mutations but had uniallelic STAT1 mutations (c.629 G>T, p.R210I; c.1073 T>G, p.L358W, c.796G>A; p.V266I; c.1154C>T, T385M [2 patients]). STAT1 phosphorylation in response to IFN-γ, IL-6, and IL-21 was increased and prolonged. CD4(+) IL-17-producing T-cell numbers were diminished. All patients had normal Treg cell percentages in the CD4(+) T-cell compartment, and their function was intact in the 2 patients tested. Patients with cells available for study had normal levels of IL-2-induced STAT5 phosphorylation. CONCLUSIONS Gain-of-function mutations in STAT1 can cause an IPEX-like phenotype with normal frequency and function of Treg cells.


Immunity | 2011

A Critical Role for STAT3 Transcription Factor Signaling in the Development and Maintenance of Human T Cell Memory

Andrea M. Siegel; Jennifer Heimall; Alexandra F. Freeman; Amy P. Hsu; Erica Brittain; Jason M. Brenchley; Gary H. Fahle; Jeffrey I. Cohen; Steven M. Holland; Joshua D. Milner

STAT3 transcription factor signaling in specific T helper cell differentiation has been well described, although the broader roles for STAT3 in lymphocyte memory are less clear. Patients with autosomal-dominant hyper-IgE syndrome (AD-HIES) carry dominant-negative STAT3 mutations and are susceptible to a variety of bacterial and fungal infections. We found that AD-HIES patients have a cell-intrinsic defect in the number of central memory CD4(+) and CD8(+) T cells compared to healthy controls. Naive T cells from AD-HIES patients had lower expression of memory-related transcription factors BCL6 and SOCS3, a primary proliferation defect, and they failed to acquire central memory-like surface phenotypes in vitro. AD-HIES patients showed a decreased ability to control varicella zoster virus (VZV) and Epstein-Barr virus (EBV) latency, and T cell memory to both of these viruses was compromised. These data point to a specific role for STAT3 in human central memory T cell formation and in control of certain chronic viruses.


Nature Genetics | 2016

Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease

Qing Zhou; Hongying Wang; Daniella M. Schwartz; Monique Stoffels; Yong Hwan Park; Yuan Zhang; Dan Yang; Erkan Demirkaya; Masaki Takeuchi; Wanxia Li Tsai; Jonathan J. Lyons; Xiaomin Yu; Claudia Ouyang; Celeste Chen; David T. Chin; Kristien Zaal; Settara C. Chandrasekharappa; Eric P. Hanson; Zhen Yu; James C. Mullikin; Sarfaraz Hasni; Ingrid E Wertz; Amanda K. Ombrello; Deborah L. Stone; Patrycja Hoffmann; Anne Jones; Beverly Barham; Helen L. Leavis; Annet van Royen-Kerkof; Cailin Sibley

Systemic autoinflammatory diseases are driven by abnormal activation of innate immunity. Herein we describe a new disease caused by high-penetrance heterozygous germline mutations in TNFAIP3, which encodes the NF-κB regulatory protein A20, in six unrelated families with early-onset systemic inflammation. The disorder resembles Behçets disease, which is typically considered a polygenic disorder with onset in early adulthood. A20 is a potent inhibitor of the NF-κB signaling pathway. Mutant, truncated A20 proteins are likely to act through haploinsufficiency because they do not exert a dominant-negative effect in overexpression experiments. Patient-derived cells show increased degradation of IκBα and nuclear translocation of the NF-κB p65 subunit together with increased expression of NF-κB–mediated proinflammatory cytokines. A20 restricts NF-κB signals via its deubiquitinase activity. In cells expressing mutant A20 protein, there is defective removal of Lys63-linked ubiquitin from TRAF6, NEMO and RIP1 after stimulation with tumor necrosis factor (TNF). NF-κB–dependent proinflammatory cytokines are potential therapeutic targets for the patients with this disease.


The Journal of Allergy and Clinical Immunology | 2013

Signal transducer and activator of transcription 1 (STAT1) gain-of-function mutations and disseminated coccidioidomycosis and histoplasmosis.

Elizabeth P. Sampaio; Amy P. Hsu; Joseph Pechacek; Hannelore I. Bax; Dalton L. Dias; Michelle L. Paulson; Prabha Chandrasekaran; Lindsey B. Rosen; Daniel Serra de Carvalho; Li Ding; Donald C. Vinh; Sarah K. Browne; Shrimati Datta; Joshua D. Milner; Douglas B. Kuhns; Debra A. Long Priel; Mohammed A. Sadat; Michael U. Shiloh; Brendan De Marco; Michael L. Alvares; Jason W. Gillman; Vivek Ramarathnam; Maria Teresa De La Morena; Liliana Bezrodnik; Ileana Moreira; Gulbu Uzel; Daniel Johnson; Christine Spalding; Christa S. Zerbe; Henry E. Wiley

BACKGROUND Impaired signaling in the IFN-γ/IL-12 pathway causes susceptibility to severe disseminated infections with mycobacteria and dimorphic yeasts. Dominant gain-of-function mutations in signal transducer and activator of transcription 1 (STAT1) have been associated with chronic mucocutaneous candidiasis. OBJECTIVE We sought to identify the molecular defect in patients with disseminated dimorphic yeast infections. METHODS PBMCs, EBV-transformed B cells, and transfected U3A cell lines were studied for IFN-γ/IL-12 pathway function. STAT1 was sequenced in probands and available relatives. Interferon-induced STAT1 phosphorylation, transcriptional responses, protein-protein interactions, target gene activation, and function were investigated. RESULTS We identified 5 patients with disseminated Coccidioides immitis or Histoplasma capsulatum with heterozygous missense mutations in the STAT1 coiled-coil or DNA-binding domains. These are dominant gain-of-function mutations causing enhanced STAT1 phosphorylation, delayed dephosphorylation, enhanced DNA binding and transactivation, and enhanced interaction with protein inhibitor of activated STAT1. The mutations caused enhanced IFN-γ-induced gene expression, but we found impaired responses to IFN-γ restimulation. CONCLUSION Gain-of-function mutations in STAT1 predispose to invasive, severe, disseminated dimorphic yeast infections, likely through aberrant regulation of IFN-γ-mediated inflammation.


Journal of Experimental Medicine | 2013

Loss-of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome

Daniel Kotlarz; Natalia Ziętara; Gulbu Uzel; Thomas Weidemann; Christian Braun; Jana Diestelhorst; Peter Krawitz; Peter N. Robinson; Jochen Hecht; Jacek Puchałka; E. Michael Gertz; Alejandro A. Schäffer; Monica G. Lawrence; Lela Kardava; Dietmar Pfeifer; Ulrich Baumann; Eva-Doreen Pfister; Eric P. Hanson; Axel Schambach; Hans Kreipe; Susan Moir; Joshua D. Milner; Petra Schwille; Stefan Mundlos; Christoph Klein

A primary immunodeficiency syndrome caused by loss-of-function mutations in the IL-21 receptor exhibits impaired B, T, and NK cell function.


Mucosal Immunology | 2012

Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection.

Nichole R. Klatt; Jacob D. Estes; Xiaoyong Sun; Alexandra M. Ortiz; John S. Barber; Levelle D. Harris; Cervasi B; Lauren K. Yokomizo; Li Pan; Carol L. Vinton; Brian Tabb; Que Dang; Vanessa M. Hirsch; Galit Alter; Yasmine Belkaid; Jeffrey D. Lifson; Guido Silvestri; Joshua D. Milner; Mirko Paiardini; Elias K. Haddad; Jason M. Brenchley

Human immunodeficiency virus (HIV) and Simian immunodeficiency virus (SIV) disease progression is associated with multifocal damage to the gastrointestinal tract epithelial barrier that correlates with microbial translocation and persistent pathological immune activation, but the underlying mechanisms remain unclear. Investigating alterations in mucosal immunity during SIV infection, we found that damage to the colonic epithelial barrier was associated with loss of multiple lineages of interleukin (IL)-17-producing lymphocytes, cells that microarray analysis showed expressed genes important for enterocyte homeostasis, including IL-22. IL-22-producing lymphocytes were also lost after SIV infection. Potentially explaining coordinate loss of these distinct populations, we also observed loss of CD103+ dendritic cells (DCs) after SIV infection, which associated with the loss of IL-17- and IL-22-producing lymphocytes. CD103+ DCs expressed genes associated with promotion of IL-17/IL-22+ cells, and coculture of CD103+ DCs and naïve T cells led to increased IL17A and RORc expression in differentiating T cells. These results reveal complex interactions between mucosal immune cell subsets providing potential mechanistic insights into mechanisms of mucosal immune dysregulation during HIV/SIV infection, and offer hints for development of novel therapeutic strategies to address this aspect of AIDS virus pathogenesis.

Collaboration


Dive into the Joshua D. Milner's collaboration.

Top Co-Authors

Avatar

Alexandra F. Freeman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven M. Holland

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jonathan J. Lyons

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kelly D. Stone

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Amy P. Hsu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nina Jones

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Thomas DiMaggio

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Celeste Nelson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Chi A. Ma

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge