Network


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

Hotspot


Dive into the research topics where Alexandra F. Freeman is active.

Publication


Featured researches published by Alexandra F. Freeman.


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 | 2009

Combined Immunodeficiency Associated with DOCK8 Mutations

Qian Zhang; Jeremiah C. Davis; Ian T. Lamborn; Alexandra F. Freeman; Huie Jing; Amanda J. Favreau; Helen F. Matthews; Joie Davis; Maria L. Turner; Gulbu Uzel; Steven M. Holland; Helen C. Su

BACKGROUND Recurrent sinopulmonary and cutaneous viral infections with elevated serum levels of IgE are features of some variants of combined immunodeficiency. The genetic causes of these variants are unknown. METHODS We collected longitudinal clinical data on 11 patients from eight families who had recurrent sinopulmonary and cutaneous viral infections. We performed comparative genomic hybridization arrays and targeted gene sequencing. Variants with predicted loss-of-expression mutations were confirmed by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay and immunoblotting. We evaluated the number and function of lymphocytes with the use of in vitro assays and flow cytometry. RESULTS Patients had recurrent otitis media, sinusitis, and pneumonias; recurrent Staphylococcus aureus skin infections with otitis externa; recurrent, severe herpes simplex virus or herpes zoster infections; extensive and persistent infections with molluscum contagiosum; and human papillomavirus infections. Most patients had severe atopy with anaphylaxis; several had squamous-cell carcinomas, and one had T-cell lymphoma-leukemia. Elevated serum IgE levels, hypereosinophilia, low numbers of T cells and B cells, low serum IgM levels, and variable IgG antibody responses were common. Expansion in vitro of activated CD8 T cells was impaired. Novel homozygous or compound heterozygous deletions and point mutations in the gene encoding the dedicator of cytokinesis 8 protein (DOCK8) led to the absence of DOCK8 protein in lymphocytes. CONCLUSIONS Autosomal recessive DOCK8 deficiency is associated with a novel variant of combined immunodeficiency.


Blood | 2011

Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome

Amy P. Hsu; Elizabeth P. Sampaio; Javed Khan; Katherine R. Calvo; Jacob Lemieux; Smita Y. Patel; David M. Frucht; Donald C. Vinh; Roger D. Auth; Alexandra F. Freeman; Kenneth N. Olivier; Gulbu Uzel; Christa S. Zerbe; Christine Spalding; Stefania Pittaluga; Mark Raffeld; Douglas B. Kuhns; Li Ding; Michelle L. Paulson; Beatriz E. Marciano; Juan Gea-Banacloche; Jordan S. Orange; Jennifer Cuellar-Rodriguez; Dennis D. Hickstein; Steven M. Holland

The syndrome of monocytopenia, B-cell and NK-cell lymphopenia, and mycobacterial, fungal, and viral infections is associated with myelodysplasia, cytogenetic abnormalities, pulmonary alveolar proteinosis, and myeloid leukemias. Both autosomal dominant and sporadic cases occur. We identified 12 distinct mutations in GATA2 affecting 20 patients and relatives with this syndrome, including recurrent missense mutations affecting the zinc finger-2 domain (R398W and T354M), suggesting dominant interference of gene function. Four discrete insertion/deletion mutations leading to frame shifts and premature termination implicate haploinsufficiency as a possible mechanism of action as well. These mutations were found in hematopoietic and somatic tissues, and several were identified in families, indicating germline transmission. Thus, GATA2 joins RUNX1 and CEBPA not only as a familial leukemia gene but also as a cause of a complex congenital immunodeficiency that evolves over decades and combines predisposition to infection and myeloid malignancy.


Blood | 2014

GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity.

Michael A. Spinner; Lauren A. Sanchez; Amy P. Hsu; Pamela A. Shaw; Christa S. Zerbe; Katherine R. Calvo; Diane C. Arthur; Wenjuan Gu; Christine M. Gould; Carmen C. Brewer; Edward W. Cowen; Alexandra F. Freeman; Kenneth N. Olivier; Gulbu Uzel; Adrian M. Zelazny; Janine Daub; Christine Spalding; Reginald J. Claypool; Neelam Giri; Blanche P. Alter; Emily M. Mace; Jordan S. Orange; Jennifer Cuellar-Rodriguez; Dennis D. Hickstein; Steven M. Holland

Haploinsufficiency of the hematopoietic transcription factor GATA2 underlies monocytopenia and mycobacterial infections; dendritic cell, monocyte, B, and natural killer (NK) lymphoid deficiency; familial myelodysplastic syndromes (MDS)/acute myeloid leukemia (AML); and Emberger syndrome (primary lymphedema with MDS). A comprehensive examination of the clinical features of GATA2 deficiency is currently lacking. We reviewed the medical records of 57 patients with GATA2 deficiency evaluated at the National Institutes of Health from January 1, 1992, to March 1, 2013, and categorized mutations as missense, null, or regulatory to identify genotype-phenotype associations. We identified a broad spectrum of disease: hematologic (MDS 84%, AML 14%, chronic myelomonocytic leukemia 8%), infectious (severe viral 70%, disseminated mycobacterial 53%, and invasive fungal infections 16%), pulmonary (diffusion 79% and ventilatory defects 63%, pulmonary alveolar proteinosis 18%, pulmonary arterial hypertension 9%), dermatologic (warts 53%, panniculitis 30%), neoplastic (human papillomavirus+ tumors 35%, Epstein-Barr virus+ tumors 4%), vascular/lymphatic (venous thrombosis 25%, lymphedema 11%), sensorineural hearing loss 76%, miscarriage 33%, and hypothyroidism 14%. Viral infections and lymphedema were more common in individuals with null mutations (P = .038 and P = .006, respectively). Monocytopenia, B, NK, and CD4 lymphocytopenia correlated with the presence of disease (P < .001). GATA2 deficiency unites susceptibility to MDS/AML, immunodeficiency, pulmonary disease, and vascular/lymphatic dysfunction. Early genetic diagnosis is critical to direct clinical management, preventive care, and family screening.


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 | 2010

Autosomal dominant and sporadic monocytopenia with susceptibility to mycobacteria, fungi, papillomaviruses, and myelodysplasia

Donald C. Vinh; Smita Y. Patel; Gulbu Uzel; Victoria L. Anderson; Alexandra F. Freeman; Kenneth N. Olivier; Christine Spalding; Stephen Hughes; Stefania Pittaluga; Mark Raffeld; Lynn Sorbara; Houda Elloumi; Douglas B. Kuhns; Maria L. Turner; Edward W. Cowen; Danielle Fink; Debra Long-Priel; Amy P. Hsu; Li Ding; Michelle L. Paulson; Adeline R. Whitney; Elizabeth P. Sampaio; David M. Frucht; Frank R. DeLeo; Steven M. Holland

We identified 18 patients with the distinct clinical phenotype of susceptibility to disseminated nontuberculous mycobacterial infections, viral infections, especially with human papillomaviruses, and fungal infections, primarily histoplasmosis, and molds. This syndrome typically had its onset in adulthood (age range, 7-60 years; mean, 31.1 years; median, 32 years) and was characterized by profound circulating monocytopenia (mean, 13.3 cells/microL; median, 14.5 cells/microL), B lymphocytopenia (mean, 9.4 cells/microL; median, 4 cells/microL), and NK lymphocytopenia (mean, 16 cells/microL; median, 5.5 cells/microL). T lymphocytes were variably affected. Despite these peripheral cytopenias, all patients had macrophages and plasma cells at sites of inflammation and normal immunoglobulin levels. Ten of these patients developed 1 or more of the following malignancies: 9 myelodysplasia/leukemia, 1 vulvar carcinoma and metastatic melanoma, 1 cervical carcinoma, 1 Bowen disease of the vulva, and 1 multiple Epstein-Barr virus(+) leiomyosarcoma. Five patients developed pulmonary alveolar proteinosis without mutations in the granulocyte-macrophage colony-stimulating factor receptor or anti-granulocyte-macrophage colony-stimulating factor autoantibodies. Among these 18 patients, 5 families had 2 generations affected, suggesting autosomal dominant transmission as well as sporadic cases. This novel clinical syndrome links susceptibility to mycobacterial, viral, and fungal infections with malignancy and can be transmitted in an autosomal dominant pattern.


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.


The Journal of Allergy and Clinical Immunology | 2010

Mutations in STAT3 and diagnostic guidelines for hyper-IgE syndrome

Cristina Woellner; E. Michael Gertz; Alejandro A. Schäffer; Macarena Lagos; Mario Perro; Erik Glocker; Maria Cristina Pietrogrande; Fausto Cossu; José Luis Franco; Nuria Matamoros; Barbara Pietrucha; Edyta Heropolitańska-Pliszka; Mehdi Yeganeh; Mostafa Moin; Teresa Espanol; Stephan Ehl; Andrew R. Gennery; Mario Abinun; Anna Bręborowicz; Tim Niehues; Sara Sebnem Kilic; Anne K. Junker; Stuart E. Turvey; Alessandro Plebani; Berta Sanchez; Ben Zion Garty; Claudio Pignata; Caterina Cancrini; Jiri Litzman; Ozden Sanal

BACKGROUND The hyper-IgE syndrome (HIES) is a primary immunodeficiency characterized by infections of the lung and skin, elevated serum IgE, and involvement of the soft and bony tissues. Recently, HIES has been associated with heterozygous dominant-negative mutations in the signal transducer and activator of transcription 3 (STAT3) and severe reductions of T(H)17 cells. OBJECTIVE To determine whether there is a correlation between the genotype and the phenotype of patients with HIES and to establish diagnostic criteria to distinguish between STAT3 mutated and STAT3 wild-type patients. METHODS We collected clinical data, determined T(H)17 cell numbers, and sequenced STAT3 in 100 patients with a strong clinical suspicion of HIES and serum IgE >1000 IU/mL. We explored diagnostic criteria by using a machine-learning approach to identify which features best predict a STAT3 mutation. RESULTS In 64 patients, we identified 31 different STAT3 mutations, 18 of which were novel. These included mutations at splice sites and outside the previously implicated DNA-binding and Src homology 2 domains. A combination of 5 clinical features predicted STAT3 mutations with 85% accuracy. T(H)17 cells were profoundly reduced in patients harboring STAT3 mutations, whereas 10 of 13 patients without mutations had low (<1%) T(H)17 cells but were distinct by markedly reduced IFN-gamma-producing CD4(+)T cells. CONCLUSION We propose the following diagnostic guidelines for STAT3-deficient HIES. Possible: IgE >1000IU/mL plus a weighted score of clinical features >30 based on recurrent pneumonia, newborn rash, pathologic bone fractures, characteristic face, and high palate. Probable: These characteristics plus lack of T(H)17 cells or a family history for definitive HIES. Definitive: These characteristics plus a dominant-negative heterozygous mutation in STAT3.


The Journal of Allergy and Clinical Immunology | 2013

Immune deficiencies, infection, and systemic immune disordersDominant 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; 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.

Collaboration


Dive into the Alexandra F. Freeman's collaboration.

Top Co-Authors

Avatar

Steven M. Holland

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

Joshua D. Milner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gulbu Uzel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Helen C. Su

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christa S. Zerbe

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joie Davis

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Victoria L. Anderson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dennis D. Hickstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge